US20090264648A1 - Synthesis of pyrazoles - Google Patents

Synthesis of pyrazoles Download PDF

Info

Publication number
US20090264648A1
US20090264648A1 US12/353,896 US35389609A US2009264648A1 US 20090264648 A1 US20090264648 A1 US 20090264648A1 US 35389609 A US35389609 A US 35389609A US 2009264648 A1 US2009264648 A1 US 2009264648A1
Authority
US
United States
Prior art keywords
compound
formula
branched
cyclic
linear
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/353,896
Inventor
Warren Chew
Zheng Wang
Gloria Cheal
Martial Bertrand
John Potoski
Mahmoud Mirmehrabi
Arianna Nencini
Riccardo Zanaletti
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Siena Biotech SpA
Wyeth LLC
Original Assignee
Siena Biotech SpA
Wyeth LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Siena Biotech SpA, Wyeth LLC filed Critical Siena Biotech SpA
Priority to US12/353,896 priority Critical patent/US20090264648A1/en
Assigned to SIENA BIOTECH S.P.A. reassignment SIENA BIOTECH S.P.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZANALETTI, RICCARDO, NENCINI, ARIANNA
Assigned to WYETH reassignment WYETH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: POTOSKI, JOHN, BERTRAND, MARTIAL, CHEAL, GLORIA, CHEW, WARREN, MIRMEHRABI, MAHMOUD, WANG, ZHENG
Publication of US20090264648A1 publication Critical patent/US20090264648A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to the synthesis of compounds with ⁇ 7 nicotinic acetylcholine receptor ( ⁇ 7 nAChR) agonistic activity, derivatives thereof, and intermediates thereto.
  • ⁇ 7 nAChR nicotinic acetylcholine receptor
  • Agents that bind to nicotinic acetylcholine receptors have been indicated as useful in the treatment and/or prophylaxis of various diseases and conditions, particularly psychotic diseases, neurodegenerative diseases involving a dysfunction of the cholinergic system, and conditions of memory and/or cognition impairment, including for example, schizophrenia, anxiety, mania, depression, manic depression, Tourette's syndrome, Parkinson's disease, Huntington's disease, cognitive disorders (such as Alzheimer's disease, Lewy Body Dementia, Amyotrophic Lateral Sclerosis, memory impairment, memory loss, cognition deficit, attention deficit, Attention Deficit Hyperactivity Disorder), and other uses such as treatment of nicotine addiction, inducing smoking cessation, treating pain (e.g.
  • the invention provides methods of preparing compounds acting as full or partial agonists at the ⁇ 7 nicotinic acetylcholine receptor ( ⁇ 7 nAChR), and intermediates thereof.
  • ⁇ 7 nAChR ⁇ 7 nicotinic acetylcholine receptor
  • Such compounds are useful for the treatment of diseases that may benefit from the activation of the alpha 7 nicotinic acetylcholine receptor such as neurological, neurodegenerative, psychiatric, cognitive, immunological, inflammatory, metabolic, addiction, nociceptive, and sexual disorders, in particular Alzheimer's disease, schizophrenia, and/or others.
  • Such compounds include those of formula I-1:
  • the present invention also provides synthetic intermediates useful for preparing such compounds.
  • the invention further provides methods to provide cost effective yields and purity.
  • FIGS. 1-7 show characterization data for hydrochloride salts.
  • FIG. 8 illustrates the effect of pH and HCl equivalence on HCl salt formation.
  • FIG. 9 shows the effects of pH and HCl equivalence on HCl salt formation
  • FIGS. 10 and 11 depict conversion of higher HCl salts to mono-HCl forms.
  • FIG. 12 shows a DSC scan of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIG. 13 shows a TGA thermogram of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIGS. 14 a - b show X-ray diffraction pattern and data for 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIG. 15 presents DVS isothermal analysis of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIG. 16 is a DSC scan of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • FIG. 17 is a TGA thermogram of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • FIGS. 18 a - b show X-ray diffraction pattern and data for 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • FIG. 19 presents a DVS isothermal analysis of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • aliphatic or “aliphatic group,” as used herein, means a straight-chain (i.e., unbranched) or branched, hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocycle” “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1-6 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms.
  • cycloaliphatic refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
  • cycloaliphatic groups include cycloalkyl and cycloalkenyl groups.
  • Suitable aliphatic groups include, but are not limited to, linear or branched alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • lower alkyl refers to a hydrocarbon chain having up to 6 carbon atoms, preferably 1 to 3 carbon atoms, and more preferably 1 to 2 carbon atoms.
  • alkyl includes, but is not limited to, straight and branched chains such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, or t-butyl.
  • alkoxy refers to the group —OR*, wherein R* is a lower alkyl group.
  • acyl refers to a group having the general formula —C( ⁇ O)R X1 , —C( ⁇ O)OR X1 , —C( ⁇ O)—O—C( ⁇ O)R X1 , —C( ⁇ O)SR X1 , —C( ⁇ O)N(R X1 ) 2 , —C( ⁇ S)R X1 , —C( ⁇ S)N(R X1 ) 2 , and —C( ⁇ S)S(R X1 ), —C( ⁇ NR X1 )R X1 , —C( ⁇ NR XI )OR XI , —C( ⁇ NR XI )SR X1 , and —C( ⁇ NR X1 )N(R X1 ) 2 , wherein R XI is hydrogen; halogen; substituted or unsubstituted hydroxyl; substituted or unsubstituted
  • acyl groups include aldehydes (—CHO), carboxylic acids (—CO 2 H), ketones, acyl halides, esters, amides, imines, carbonates, carbamates, and ureas.
  • Acyl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyl
  • halogen or “halo,” as used herein, refer to chlorine, bromine, fluorine or iodine.
  • alkenyl refers to an aliphatic straight or branched hydrocarbon chain having 2 to 4 carbon atoms that has one or more double bonds.
  • alkenyl groups include vinyl, prop-1-enyl, allyl, methallyl, but-1-enyl, but-2-enyl, or but-3-enyl.
  • lower alkenyl refers to an alkenyl group having 1 to 3 carbon atoms.
  • aryl refers to phenyl or an 8-10 membered bicyclic partially unsaturated or aryl ring.
  • exemplary aryl groups include phenyl and naphthyl.
  • aryl refers to an 8-10 membered bicyclic partially unsaturated the wherein at least one of the rings is aromatic.
  • heteroaryl refers to a 5-6 membered monocyclic heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic partially unsaturated or heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • heteroaryls include, but are not limited to, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, indazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, isobenzothienyl, quinolyl, isoquinolyl, quinoxalinyl, or quinazolinyl.
  • compounds of the invention may contain “optionally substituted” moieties.
  • substituted whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group are independently halogen; —(CH 2 ) 0-4 R ⁇ ; —(CH 2 ) 0-4 OR ⁇ ; —O(CH 2 ) 0-4 R ⁇ , —O—(CH 2 ) 0-4 C(O)OR ⁇ ; —(CH 2 ) 0-4 CH(OR ⁇ ) 2 ; —(CH 2 ) 0-4 SR ⁇ ; —(CH 2 ) 0-4 Ph, which may be substituted with R ⁇ ; —(CH 2 ) 0-4 O(CH 2 ) 0-1 Ph which may be substituted with R ⁇ ; —CH ⁇ CHPh, which may be substituted with R ⁇ ; —(CH 2 ) 0-4 O(CH 2 ) 0-1 -pyridyl which may be substituted with R ⁇ ; —NO 2 ; —CN;
  • Suitable monovalent substituents on R ⁇ are independently halogen, —(CH 2 ) 0-2 R ⁇ , -(haloR ⁇ ), —(CH 2 ) 0-2 OH, —(CH 2 ) 0-2 OR ⁇ , —(CH 2 ) 0-2 CH(OR ⁇ ) 2 ; —O(haloR ⁇ ), —CN, —N 3 , —(CH 2 ) 0-2 C(O)R ⁇ , —(CH 2 ) 0-2 C(O)OH, —(CH 2 ) 0-2 C(O)OR ⁇ , —(CH 2 ) 0-2 SR ⁇ , —(CH 2 ) 0-2 SH, —(CH 2 ) 0-2 NH 2 , —(CH 2 ) 0-2 NHR ⁇ , —(CH 2
  • Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: ⁇ O, ⁇ S, ⁇ NNR* 2 , ⁇ NNHC(O)R*, ⁇ NNHC(O)OR*, ⁇ NNHS(O) 2 R*, ⁇ NR*, ⁇ NOR*, —(C(R* 2 )) 2-3 O—, or S(C(R* 2 )) 2-3 S—, wherein each independent occurrence of R* is selected from hydrogen, C 1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: —O(CR* 2 ) 2-3 O—, wherein each independent occurrence of R* is selected from hydrogen, C 1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R* include halogen, —R ⁇ , -(haloR ⁇ ), —OH, —OR ⁇ , —O(haloR ⁇ ), —CN, —C(O)OH, —C(O)OR ⁇ , —NH 2 , —NHR ⁇ , —NR ⁇ 2 , or —NO 2 , wherein each R ⁇ is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1-4 aliphatic, —CH 2 Ph, —O(CH 2 ) 0-1 Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include —R ⁇ , —NR ⁇ 2 , —C(O)R ⁇ , —C(O)OR ⁇ , —C(O)C(O)R ⁇ , —C(O)CH 2 C(O)R ⁇ , —S(O) 2 R ⁇ , —S(O) 2 NR ⁇ 2 , —C(S)NR ⁇ 2 , —C(NH)NR ⁇ 2 , or —N(R ⁇ )S(O) 2 R ⁇ ; wherein each R ⁇ is independently hydrogen, C 1-6 aliphatic which may be substituted as defined below, unsubstituted —OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrence
  • Suitable substituents on the aliphatic group of R ⁇ are independently halogen, —R ⁇ , -(haloR ⁇ ), —OH, —OR ⁇ , —O(haloR ⁇ ), —CN, —C(O)OH, —C(O)OR ⁇ , —NH 2 , —NHR ⁇ , —NR ⁇ 2 , or —NO 2 , wherein each R ⁇ is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1-4 aliphatic, —CH 2 Ph, —O(CH 2 ) 0-1 Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • pharmaceutically acceptable salts or “pharmaceutically acceptable salt” includes acid addition salts, that is salts derived from treating compounds of formulae I-1 and A with an organic or inorganic acid such as, for example, acetic, lactic, citric, cinnamic, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, oxalic, propionic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, glycolic, pyruvic, methanesulfonic, ethanesulfonic, toluenesulfonic, salicylic, benzoic, or similarly known acceptable acids.
  • a compound of formulae I-1 and A contains a substituent with acidic properties, for instance, phenolic hydroxyl, —SO 2 H or —CO 2 H
  • the term also includes salts derived from bases, for example, sodium salts.
  • the present invention provides a method for preparing compounds of formula I-1:
  • the present compounds are generally prepared according to Scheme 1 set forth below:
  • Ring A, Y, Y′, T, and Ar is defined in formula I-1 and described in classes and subclasses above and herein, and LG 2 and LG 3 are leaving groups.
  • the T moiety on formulae I-1, B′, and F′ is a C 3-5 bivalent hydrocarbon chain, optionally substituted with one or more halogen, hydroxy, C 1-5 alkyl, alkoxy, fluoroalkyl, hydroxyalkyl, alkylidene, or fluoroalkylidene groups.
  • the T moiety is a C 4 bilvalent hydrocarbon chain.
  • Y is a nitrogen atom. In certain embodiments, Y′ is a nitrogen atom.
  • Ring A of a compound of formula G′ is a 4-8 membered saturated ring.
  • a compound of formula G′ is a piperazine derivative.
  • a compound of formula G′ is N-acetylhomopiperazine.
  • Ar is an optionally substituted group selected from 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. In some embodiments, Ar is an optionally substituted 6 membered aryl group. In certain embodiments, Ar is methoxy-phenyl. In certain embodiments, Ar is 4-methoxy-phenyl. In some embodiments, Ar is hydroxy-phenyl. In some embodiments, Ar is a phenyl group substituted with a —OSO 3 H group.
  • a compound of formula C′ is reacted in a suitable solvent, as defined and described herein, with a compound of formula F′ to form a compound of formula B′.
  • the LG 2 group of formulae B′ and F′ is a suitable leaving group, as defined and described herein.
  • the LG 2 group of formulae B′ and F′ is halogen, —OMs, —OTs, or —OTf.
  • the LG 2 group of formulae B′ and F′ is —Br.
  • the LG 3 group of formula F′ is a suitable leaving group, as defined and described herein.
  • the LG 3 group of formula F′ is halogen, —OR,
  • each R is independently hydrogen or an optionally substituted group selected from C 1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • LG 3 groups of formula F′ include —OH, —OMe, —OEt, —Cl, —Br,
  • LG 3 is —Cl.
  • LG 3 is —OH
  • the reaction between a compound of formula F′ and a compound of formula B′ is carried out using suitable peptide coupling conditions.
  • suitable peptide coupling conditions are well known in the art and include those described in detail in Han et al., Tetrahedron, 60, 2447-67 (2004), the entirety of which is hereby incorporated by reference.
  • the peptide coupling conditions include the addition of HOBt, DMAP, BOP, HBTU, HATU, BOMI, DCC, EDC, IBCF, or a combination thereof.
  • the compound of formula F′ is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F′ is 5-bromovaleryl chloride.
  • Suitable solvents for step S-1 include aprotic solvents, aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, aliphatic ethers or combinations thereof.
  • the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof.
  • the solvent is a mixture of acetonitrile and DMF.
  • the solvent is a 9:1 mixture of acetonitrile:DMF.
  • Suitable bases for step S-1 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof.
  • the base is diisopropylethylamine.
  • Suitable temperatures at which the reaction described in step S-1 may occur include about ⁇ 20° C. to about 60° C. In certain embodiments, the temperature is about ⁇ 10° C. to about 25° C. In some embodiments, the temperature is about ⁇ 10° C.
  • a compound of formula F′ is added slowly to a compound of formula C′.
  • reaction conditions may result in the formation of a regioisomer in step S-1 (i.e., reaction of endocyclic pyrazole nitrogen with a compound of formula F′).
  • the product of step S-1 is treated to remove the undesired regioisomer.
  • the product of step S-1 is re-slurried with a particular solvent or solvents to remove the regioisomer.
  • the product of step S-1 is re-slurried with t-butyl methyl ether to remove the regioisomer.
  • a compound of formula B′ is reacted in a suitable solvent with a compound of formula G′, optionally in the presence of a suitable base and/or cataylst, to produce compound I-1.
  • a compound of formula B′ is 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide
  • 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom.
  • iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof. While not wishing to be bound by any particular theory, it is believed that 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be a more reactive species in step S-2 than 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, due to the greater leaving group character of iodide over bromide.
  • Suitable solvents for step S-2 include aprotic solvents, aliphatic halides, aliphatic ethers or combinations thereof.
  • the solvent is selected from dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof.
  • the solvent is acetone.
  • Suitable bases for step S-2 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof.
  • the base is potassium carbonate.
  • the base is diisopropylethylamine.
  • the iodide source used in step S-2 is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B′. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B′.
  • Suitable temperatures at which the reaction described in step S-2 may occur include about ⁇ 20° C. to about 60° C. In certain embodiments, the temperature is about ⁇ 10° C. to about 25° C. In some embodiments, the temperature is about 25° C.
  • a compound of formula I-1 may be further transformed into a pharmaceutically acceptable salt.
  • the pharmaceutically acceptable salt is a hydrochloride salt.
  • the pharmaceutically acceptable salt is a mono hydrochloride salt.
  • an extractive workup step may be performed following step S-2.
  • the organic phase comprises one or more organic solvents.
  • the organic solvents are ethanol, methyl tetrahydrofuran, dichloromethane, or a combination thereof.
  • the solvents are ethanol and methyl tetrahydrofuran.
  • the solvents are ethanol and dichloromethane.
  • the organic solvents are 5% ethanol in methyl tetrahydrofuran.
  • the organic solvents are 5% ethanol in dichloromethane.
  • dimer compounds may be formed as side products at step S-2. In such cases, it has been found that performing additional washes following HCl salt formation may be useful for reducing the presence of such dimer products.
  • each of the aforementioned synthetic steps may be performed sequentially with isolation of each intermediate B′ and I-1 performed after each step.
  • each of steps S-1 and S-2, as depicted in Scheme I above, as well as any subsequent salt formation may be performed in a manner whereby no isolation of one or more intermediates B′ and I-1 is performed.
  • steps S-1 and S-2 are performed in sequence without any isolation of intermediates.
  • step S-2 and salt formation are performed without any isolation of intermediates. While not wishing to be bound by any particular theory, it is believed that such techniques may be useful in obtaining compounds of formulae B′, I-1, and pharmaceutically acceptable salts thereof in greater yield and purity.
  • the present invention provides a method for preparing compound 1,5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrogen chloride:
  • Compound I a potent ⁇ 7 nAChR agonist, is effective in treating diseases that may benefit from the activation of the alpha 7 nicotinic acetylcholine receptor such as neurological, neurodegenerative, psychiatric, cognitive, immunological, inflammatory, metabolic, addiction, nociceptive, and sexual disorders, in particular Alzheimer's disease, schizophrenia, and/or others.
  • the present compounds are generally prepared according to Scheme 2 set forth below:
  • the present invention provides methods for preparing a free base, A, according to the steps depicted in Scheme 2, above.
  • an aromatic ester of formula E is reacted, optionally in a suitable solvent, with a suitable base and acetonitrile to provide ⁇ -ketonitrile D.
  • suitable bases include metal alkoxides and metal hydrides.
  • the base is an alkali hydride, a metal alkyl, a metal amide, or a metal silazide.
  • Suitable bases include KH, n-butyl lithium, hexyl lithium, lithium diisopropylamide, Li—N(Si-alkyl) 2 , lithium hexamethyldisilazide (LiHMDS), sodium hexamethyldisilazide (NaHMDS), potassium hexamethyldisilazide (KHMDS) potassium t-amylate, sodium t-butoxide (NaOtBu), and sodium hydride (NaH).
  • the base is LiHMDS.
  • the acetonitrile used in step S-3 may be used in a range of equivalents relative to aromatic ester E. In certain embodiments, the equivalents of acetonitrile range from 0.5 to 50 equivalents. In certain embodiments, the equivalents of acetonitrile range from 2 to 10 equivalents. In some embodiments, the equivalents of acetonitrile range from 4 to 6 equivalents.
  • Suitable temperatures at which the reaction described in step S-3 may occur include about ⁇ 20° C. to about 80° C. In certain embodiments, the temperature is about ⁇ 20° C. to about 0° C. In some embodiments, the temperature is about ⁇ 10° C.
  • Step S-3 may optionally employ a suitable solvent.
  • a suitable solvent is a solvent or a solvent mixture that, in combination with the combined reacting partners and reagents, facilitates the progress and/or rate of the reaction.
  • the suitable solvent may solubilize one or more of the reaction components, or, alternatively, the suitable solvent may facilitate the suspension of one or more of the reaction components; see, generally, “Advanced Organic Chemistry,” Jerry March, 5 th edition, John Wiley and Sons, N.Y.
  • solvents suitable for use in step S-3 include anhydrous aprotic solvents, such as aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, aliphatic nitriles, and aliphatic ethers.
  • the solvent is selected from toluene, acetonitrile, diethyl ether, t-butyl methyl ether, THF, benzene, dichloromethane or combinations thereof. In certain embodiments, no solvent is used.
  • the LG 1 group of formula E is a suitable leaving group.
  • a suitable leaving group is a chemical group that is readily displaced by a desired incoming chemical moiety.
  • Suitable leaving groups are well known in the art, e.g., see, “Advanced Organic Chemistry,” Jerry March, 5 th Ed., pp. 351-357, John Wiley and Sons, N.Y.
  • Such leaving groups include, but are not limited to, halogen, alkoxy, sulphonyloxy, optionally substituted alkylsulphonyl, optionally substituted alkenylsulfonyl, optionally substituted arylsulfonyl, and diazonium moieties.
  • Examples of some suitable leaving groups include chloro, iodo, bromo, fluoro, methanesulfonyl (mesyl), tosyl, triflate, nitro-phenylsulfonyl (nosyl), and bromo-phenylsulfonyl (brosyl).
  • the LG 1 group of formula E is halogen, —OR,
  • each R is independently hydrogen or an optionally substituted group selected from C 1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • LG 1 groups of formula E include —OH, —OMe, —OEt, Cl, Br,
  • LG 1 is —OR, wherein R is C 1-6 alkyl. In certain embodiments, LG 1 is —OR, wherein R is methyl.
  • the suitable leaving group may be generated in situ within the reaction medium.
  • a leaving group may be generated in situ from a precursor of that compound wherein said precursor contains a group readily replaced by said leaving group in situ.
  • ⁇ -ketonitrile D is reacted in a suitable solvent with hydrazine, or an equivalent thereof, to form aryl aminopyrazole C.
  • hydrazine equivalents are well known to one of ordinary skill in the art and include, but are not limited to, anhydrates, hydrates, monohydrates, monohydrochlorides, dihydrochlorides, and sulfates.
  • the hydrazine equivalent used in step S-4 is a hydrate.
  • the hydrazine equivalent is a monohydrate.
  • Suitable solvents for step S-4 include alkyl alcohols, such as C 1 to C 4 alcohols (e.g. ethanol, methanol, 2-propanol), aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, or aliphatic ethers or combinations thereof.
  • the solvent is selected from ethanol, toluene, dichloromethane, diethyl ether, THF, benzene or combination thereof.
  • the solvent is ethanol.
  • Suitable temperatures at which the reaction described in step S-4 may occur include about 10° C. to about 150° C. In certain embodiments, the temperature is about 30° C. to about 70° C. In some embodiments, the temperature is about 60° C.
  • aryl aminopyrazole C is reacted in a suitable solvent with a compound of formula F to form a compound of formula B.
  • the LG 2 group of formulae B and F is a suitable leaving group, as defined and described herein.
  • the LG 2 group of formulae B and F is halogen, —OMs, —OTs, or —OTf.
  • Examples of the LG 2 group of formulae B and F include —Br, —I, —OMs, —OTs, and —OTf.
  • the LG 3 group of formula F is a suitable leaving group, as defined and described herein.
  • the LG 3 group of formula F is halogen, —OR,
  • each R is independently hydrogen or an optionally substituted group selected from C 1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • LG 3 groups of formula F include —OH, —OMe, —OEt, —Cl, —Br,
  • LG 3 is —Cl.
  • LG 3 is —OH
  • the reaction between a compound of formula F and a compound of formula B is carried out using suitable peptide coupling conditions.
  • suitable peptide coupling conditions are well known in the art and include those described in detail in Han et al., Tetrahedron, 60, 2447-67 (2004), the entirety of which is hereby incorporated by reference.
  • the peptide coupling conditions include the addition of HOBt, DMAP, BOP, HBTU, HATU, BOMI, DCC, EDC, IBCF, or a combination thereof.
  • the compound of formula F is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F is 5-bromovaleryl chloride.
  • Suitable solvents for step S-5 include aprotic solvents, aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, aliphatic ethers or combinations thereof.
  • the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof.
  • the solvent is a mixture of acetonitrile and DMF.
  • the solvent is a 9:1 mixture of acetonitrile:DMF.
  • Suitable bases for step S-5 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof.
  • the base is diisopropylethylamine.
  • Suitable temperatures at which the reaction described in step S-5 may occur include about ⁇ 20° C. to about 60° C. In certain embodiments, the temperature is about ⁇ 10° C. to about 25° C. In some embodiments, the temperature is about ⁇ 10° C.
  • a compound of formula F is added slowly to a compound of formula C.
  • reaction conditions may result in the formation of a regioisomer in step S-5 (i.e., reaction of endocyclic pyrazole nitrogen with a compound of formula F).
  • the product of step S-5 is treated to remove the undesired regioisomer.
  • the product of step S-5 is re-slurried with a particular solvent or solvents to remove the regioisomer.
  • the product of step S-5 is re-slurried with t-butyl methyl ether to remove the regioisomer.
  • a compound of formula B is reacted in a suitable solvent with N-acetylhomopiperazine, optionally in the presence of a suitable base and/or catalyst, to produce compound A, 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide.
  • 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom.
  • iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof.
  • 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be a more reactive species in step S-6 than 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, due to the greater leaving group character of iodide over bromide.
  • Suitable solvents for step S-6 include aprotic solvents, aliphatic halides, aliphatic ethers or combinations thereof.
  • the solvent is selected from dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof.
  • the solvent is acetone.
  • Suitable bases for step S-6 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof.
  • the base is potassium carbonate.
  • the potassium carbonate is milled.
  • the base is diisopropylethylamine.
  • the iodide source used in step S-6 is sodium iodide.
  • the iodide source is potassium iodide.
  • the potassium iodide is milled.
  • the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B.
  • Suitable temperatures at which the reaction described in step S-6 may occur include about ⁇ 20° C. to about 60° C. In certain embodiments, the temperature is about ⁇ 10° C. to about 25° C. In some embodiments, the temperature is about 25° C.
  • an extractive workup step may be performed following step S-6.
  • the organic phase comprises one or more organic solvents.
  • the organic solvents are ethanol, methyl tetrahydrofuran, dichloromethane, or a combination thereof.
  • the solvents are ethanol and methyl tetrahydrofuran.
  • the solvents are ethanol and dichloromethane.
  • the organic solvents are 5% ethanol in methyl tetrahydrofuran.
  • the organic solvents are 5% ethanol in dichloromethane.
  • dimer compounds may be formed as side products at step S-6. In such cases, it has been found that performing additional washes following HCl salt formation may be useful for reducing the presence of such dimer products.
  • step S-7 compound A is reacted with hydrogen chloride, or an equivalent thereof, to form compound 1,5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrogen chloride.
  • Suitable solvents for step S-7 include polar solvents such as C 1 to C 4 alcohols (e.g. ethanol, methanol, 2-propanol), water, acetone or combinations thereof.
  • the solvent is selected from ethanol, water, acetone, or combination thereof.
  • the solvent a mixture of acetone, water, and ethanol.
  • the ethanol is ethanol 1 L (denatured with 9% acetone). In some embodiments, the ethanol is ethanol 2B (denatured with 0.5% toluene).
  • step S-7 a number of suitable forms of hydrogen chloride can be used in step S-7 to produce the desired hydrochloride salt.
  • suitable forms include hydrogen chloride gas, aqueous solutions of hydrogen chloride, and solutions of hydrogen chloride in aliphatic ethers.
  • the HCl is provided as an aqueous solution in acetone.
  • the number of equivalents of HCl used relative to compound of formula A is about 0.5-1.1 equivalents. In certain embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.8-1.0 equivalents. In some embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.93 equivalents.
  • step S-7 may result in the formation of a di-HCl salt.
  • formation of such di-HCl salts is minimized or avoided by the addition of HCl in equivalents as described herein.
  • formation of such di-HCl salts is minimized or avoided by careful, controlled addition of HCl.
  • Suitable temperatures at which the reaction described in step S-7 may occur include about ⁇ 20° C. to about 60° C. In certain embodiments, the temperature is about ⁇ 10° C. to about 35° C. In some embodiments, the temperature is about 25° C. to about 30° C.
  • each of the aforementioned synthetic steps may be performed sequentially with isolation of each intermediate D, C, B, and A performed after each step.
  • each of steps S-3, S-4, S-5, S-6 and S-7, as depicted in Scheme 2 above may be performed in a manner whereby no isolation of one or more intermediates D, C, B, and A is performed.
  • steps S-5, S-6, and S-7 are performed in sequence without any isolation of intermediates.
  • steps S-6 and S-7 are performed in sequence without any isolation of intermediates. While not wishing to be bound by any particular theory, it is believed that such techniques may be useful in obtaining compounds of formulae B, A, and I in greater yield and purity.
  • compound A can be prepared according to Scheme 3 set forth below:
  • an ester of formula G is coupled with N-acetylhomopiperazine to form an ester of formula H.
  • R is optionally substituted group selected from C 1-6 aliphatic. In some embodiments, R is C 1-3 aliphatic. In some embodiments, R is methyl.
  • a suitable base may be used to facilitate the reaction between a compound of formula G and N-acetylhomopiperazine.
  • bases include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, potassium carbonate, and combinations thereof.
  • the base is potassium carbonate.
  • the iodo analog may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom.
  • iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof.
  • the iodide source is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula G. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula G.
  • step S-3a is carried out in the presence of a suitable solvent.
  • the solvent is selected from a polar aprotic solvent.
  • Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetamide, acetone, or combination thereof.
  • the solvent is acetone.
  • an ester of formula H is saponified with a suitable acid or base to provide carboxylic acid J.
  • suitable bases include strong inorganic bases i.e., those that completely dissociate in water under formation of hydroxide anion. Examples of such bases include alkaline metals, alkaline earth metal hydroxides, and combinations thereof.
  • a suitable acid is a Lewis acid.
  • step S-5a carboxylic acid J is chlorinated to form acyl chloride K.
  • the chlorination is facilitated by POCl 3 .
  • step S-5a is carried out in the presence of a suitable solvent.
  • the solvent is selected from a polar aprotic solvent.
  • Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetamide, acetone, or combination thereof.
  • the solvent is dimethylacetamide.
  • acyl chloride K is reacted with aryl aminopyrazole C to provide compound A.
  • Compound A may then be used as described above and herein.
  • the present invention provides a method for preparing compound I-1′:
  • suitable forms of hydrogen chloride can be used to produce the desired hydrochloride salt.
  • suitable forms include hydrogen chloride gas, aqueous solutions of hydrogen chloride, and solutions of hydrogen chloride in aliphatic ethers.
  • the HCl is provided as an aqueous solution in acetone.
  • compound A is treated with hydrochloric acid in a solvent mixture of acetone, water, and ethanol.
  • the number of equivalents of HCl used relative to compound of formula I-1 is about 0.5-1.1 equivalents. In certain embodiments, the number of equivalents of HCl used relative to compound of formula I-1 is about 0.8-1.0 equivalents. In some embodiments, the number of equivalents of HCl used relative to compound of formula I-1 is about 0.93 equivalents.
  • the present invention provides a method for preparing compound I-1:
  • LG 2 is a suitable leaving group
  • the LG 2 group of formula B′ is a suitable leaving group, as defined and described herein. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. Examples of the LG 2 group of formula B′ include Br, I, —OMs, —OTs, and —OTf.
  • 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom.
  • iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof.
  • the iodide source is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B′. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B′.
  • a suitable base may be used to facilitate the reaction between a compound of formula B′ and a compound of formula G′.
  • bases include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, potassium carbonate, and combinations thereof.
  • the base is diisopropylethylamine.
  • the base is potassium carbonate.
  • the transformation of a compound of formula B′ to compound I-1 is performed in the presence of a suitable solvent.
  • the solvent is selected from a polar aprotic solvent.
  • Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof.
  • the solvent is acetone.
  • the present invention provides a method for preparing a compound of formula B′:
  • LG 3 is a suitable leaving group, to form a compound of formula B′.
  • the LG 2 group of formulae B′ and F′ is a suitable leaving group, as defined and described herein. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. In certain embodiments, the LG 2 group of formulae B′ and F′ include Br, I, —OMs, —OTs, and —OTf.
  • LG 3 group of formula F′ is a suitable leaving group, and defined and described herein.
  • Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OR,
  • each R is independently hydrogen or an optionally substituted group selected from C 1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • LG 3 groups of formula F′ include —OH, —OMe, —OEt, —Cl, —Br,
  • LG 3 is —Cl.
  • the compound of formula F′ is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F′ is 5-bromovaleryl chloride.
  • Suitable bases to facilitate the transformation of compound C′ to a compound of formula B′ include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, and combinations thereof.
  • the base is diisopropylethylamine.
  • the transformation of compound C′ to a compound of formula B′ is carried out in the presence of a suitable solvent.
  • the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof.
  • the solvent is a mixture of acetonitrile and DMF.
  • the solvent is a 9:1 mixture of acetonitrile:DMF.
  • the present invention provides a method for preparing compound I:
  • suitable forms of hydrogen chloride can be used to produce the desired hydrochloride salt.
  • suitable forms include hydrogen chloride gas, aqueous solutions of hydrogen chloride, and solutions of hydrogen chloride in aliphatic ethers.
  • the HCl is provided as an aqueous solution in acetone.
  • compound A is treated with hydrochloric acid in a solvent mixture of acetone, water, and ethanol.
  • the number of equivalents of HCl used relative to compound of formula A is about 0.5-1.1 equivalents. In certain embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.8-1.0 equivalents. In some embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.93 equivalents.
  • the present invention provides a method for preparing compound A:
  • LG 2 is a suitable leaving group
  • the LG 2 group of formula B is a suitable leaving group. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. Examples of the LG 2 group of formula B include —Br, —I, —OMs, —OTs, and —OTf.
  • 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom.
  • iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof.
  • the iodide source is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B.
  • a suitable base may be used to facilitate the reaction between a compound of formula B and N-acetylhompiperazine.
  • bases include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, potassium carbonate, and combinations thereof.
  • the base is diisopropylethylamine.
  • the base is potassium carbonate.
  • the transformation of a compound of formula B to compound A is performed in the presence of a suitable solvent.
  • the solvent is selected from a polar aprotic solvent.
  • Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof.
  • the solvent is acetone.
  • the present invention provides a method for preparing a compound of formula B:
  • the LG 2 group of formulae B and F is a suitable leaving group. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. Examples of the LG 2 group of formulae B and F include —Br, —I, —OMs, —OTs, and —OTf.
  • the LG 3 group of formula F is a suitable leaving group.
  • Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OR,
  • each R is independently hydrogen or an optionally substituted group selected from C 1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • LG 3 groups of formula F include —OH, —OMe, —OEt, —Cl, —Br,
  • LG 3 is —Cl.
  • the compound of formula F is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F is 5-bromovaleryl chloride.
  • Suitable bases to facilitate the transformation of compound C to a compound of formula B include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, and combinations thereof.
  • the base is diisopropylethylamine.
  • the transformation of compound C to a compound of formula B is carried out in the presence of a suitable solvent.
  • the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof.
  • the solvent is a mixture of acetonitrile and DMF.
  • the solvent is a 9:1 mixture of acetonitrile:DMF.
  • the present invention provides a method for preparing compound C:
  • the hydrazine equivalent used is a hydrate. In some embodiments, the hydrazine equivalent is a monohydrate.
  • the transformation of compound D to compound C is carried out in the presence of a suitable solvent.
  • the solvent is selected from ethanol, toluene, dichloromethane, diethyl ether, THF, benzene or combination thereof. In some embodiments, the solvent is ethanol.
  • the present invention provides a method for preparing compound D:
  • LG 1 is a leaving group, with acetonitrile to form a mixture thereof, and; (b) treating said mixture with a suitable base to give compound D.
  • the base is selected the group consisting of NaH, LDA, NaHMDS, LHMDS, KHMDS, potassium t-amylate, BuLi, and NaOtBu. In some embodiments, the base is LHMDS.
  • the acetonitrile used in step (a) may be used in a range of equivalents relative to aromatic ester E. In certain embodiments, the equivalents of acetonitrile range from 0.5 to 50 equivalents. In certain embodiments, the equivalents of acetonitrile range from 2 to 10 equivalents. In some embodiments, the equivalents of acetonitrile range from 4 to 6 equivalents.
  • the transformation of a compound of formula E to compound D may be carried out in the presence of a suitable solvent.
  • the solvent is selected from toluene, acetonitrile, diethyl ether, t-butyl methyl ether, THF, benzene, dichloromethane or combinations thereof. In certain embodiments, no solvent is used.
  • the present invention provides compound I substantially free of N-acetylhompiperazine, 5-iodovaleryl chloride, 5-bromovaleryl chloride, 5-chlorovaleryl chloride, and compounds F-1, F-2, and F-3:
  • Compounds F-1, F-2, and F-3 are impurities that may arise from steps S-1, S-2, S-5, or S-6 as described above and herein. “Substantially free,” as used herein, means that at least about 80% by weight of the desired compound is present. In other embodiments, at least about 92% by weight of a desired compound is present. In still other embodiments of the invention, at least about 99% by weight of a desired compound is present.
  • Such impurities may be isolated from product mixtures by any method known to those skilled in the art, including liquid chromatography (LC).
  • the present invention provides compound I having total impurities of less than 0.5%, less than 0.4%, or less than 0.3% by weight.
  • the present invention provides methods that provide compound I in substantially higher yields than described previously (U.S. Patent Application Ser. No. 60/880,629, filed Jan. 16, 2007).
  • Compounds of the invention can be in the form of free bases or acid addition salts, preferably salts with pharmaceutically acceptable acids.
  • Pharmacological activity of a representative group of compounds of formula I-1 was demonstrated in an in vitro assay utilizing cells stably transfected with the alpha 7 nicotinic acetylcholine receptor and cells expressing the alpha 1 and alpha 3 nicotinic acetylcholine receptors and 5HT 3 receptor as controls for selectivity.
  • prodrugs of compounds of formula I-1 may be provided according to the present invention in any of a variety of useful forms, for example as pharmaceutically acceptable salts, as particular crystal forms, etc.
  • prodrugs of compounds of formula I-1 are provided.
  • Various forms of prodrugs are known in the art, for example as discussed in Bundgaard (ed.), Design of Prodrugs , Elsevier (1985); Widder et al. (ed.), Methods in Enzymology , vol. 4, Academic Press (1985); Kgrogsgaard-Larsen et al.
  • Preparative HLPC was run using a Waters 2767 system with a binary Gradient Module Waters 2525 pump and coupled to a Waters Micromass ZQ (ES) or Waters 2487 DAD, using a Supelco Discovery HS C18 5.0 ⁇ m 10 ⁇ 21.2 mm column Gradients were run using 0.1% formic acid/water and 0.1% formic acid/acetonitrile with gradient 5/95 to 95/5 in the run time indicated in the Examples.
  • Aryl or heteroaryl methyl carboxylate were commercially available or were synthesized according to the following standard procedure: the aryl or heteroaryl carboxylic acid (32 mmol) was dissolved in MeOH (40 mL) and sulfuric acid (1 mL) was added. The mixture was refluxed overnight, after which the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated aqueous NaHCO3 solution. The organic phase was dried and evaporated under reduced pressure, and the crude was used without further purification.
  • the reaction was then allowed to cool down to room temperature and the solid formed was filtered and then dissolved in water.
  • the solution was then acidified with 2N HCl solution and at pH between 2-6 (depending on the ring substitution on the aryl/heteroaryl system) the product precipitated and was filtered off. If no precipitation occurred, the product was extracted with DCM.
  • Aryl- or heteroaryl-carboxylic acid methyl esters are commercially available or were synthesized under the standard procedure, as described in general procedure A1.
  • Yields were generally between 65 and 90%.
  • POC 3 (2 eq with respect to the aryl/heteroaryl acetophenone) were added dropwise to 4 molar equivalents of anhydrous DMF cooled down to 0° C., at such a rate that the temperature did not exceed 10° C.
  • the acetophenone (1 eq) was then added dropwise and the reaction was allowed to reach room temperature.
  • the reaction was then stirred for further 30 min and then 0.4 mmol of hydroxylamine hydrochloride were added.
  • the reaction was then heated up to 50° C., after which heating was removed and additional 4 eq. of hydroxylamine hydrochloride were added portionwise (at such a rate that the temperature never exceeded 120° C.).
  • the reaction was then stirred until the temperature of the mixture spontaneously decreased to 25° C.
  • Water (100 mL) were then added and the mixture was extracted with diethyl ether. The organic phase was dried over Na 2 SO 4 and concentrated under reduced pressure. The crude product was used for the next step without further purification.
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 2-trifluoromethyl-benzoic acid methyl ester (3.1 g, 14.0 mmol, 1.0 eq). The crude was precipitated from HCl to give the title product as a yellow solid (2.8 g, yield: 94%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude was purified through Si column (eluent: DCM) and dried to give the title product (0.6 g, 20% Yield).
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1), refluxing the mixture overnight and then for 2 h at 110° C.
  • the crude product was extracted with DCM and used in the following step without further purification (2.2 g, yield: 76%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude was purified through Si column (eluent: DCM) and washed with water, extracted and dried to give the title product (0.25 g, yield 10%).
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 2-chloro-4-fluoro-benzoic acid methyl ester (0.7 g, 3.7 mmol, 1.0 eq).
  • the crude product was extracted with DCM and used in the following step without further purification (0.4 g, yield: 60%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude was dissolved in DCM, washed with sat NaHCO 3 , extracted and dried to give the title product (0.12 g, yield 26%).
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 5-tert-Butyl-thiophene-2-carboxylic acid methyl ester (3.0 g, 15.0 mmol, 1.0 eq).
  • the crude product was extracted with DCM and used in the following step without further purification (2.7 g, yield: 86%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude was washed with water and precipitated to give the title product (2.7 g, yield 91%).
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 3-Chloro-2-methyl-benzoic acid methyl ester (3.1 g, 16.8 mmol, 1.0 eq).
  • the crude product was precipitated form water and used in the following step without further purification (2.4 g, yield: 74%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column (20 g) with gradient elution from 100% EtOAc to EtOAc-MeOH 80:20.
  • the title product (1.3 g, yield 50%) was obtained.
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 2-ethyl-benzoic acid methyl ester (2.9 g, 17.6 mmol, 1.0 eq).
  • the crude product was extracted with DCM as a yellow oil and used in the following step without further purification (2.8 g, yield: 92%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column (20 g) with gradient elution from 100% EtOAc to EtOAc-MeOH 80:20.
  • the title product (1.2 g, yield 40%) was obtained
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 4-methoxy-benzoic acid methyl ester (3.0 mL, 18.0 mmol, 1.0 eq), NaH (1.4 g, 36.0 mmol, 2.0 eq) and propionitrile (6.1 mL, 84.9 mmol, 4.7 eq).
  • the crude was purified through Si-column (eluent hexane/ethyl acetate) to give 2.1 g of title product (yield: 62%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was washed with basic water and dried, and the title product (1.8 g, yield 80%) was used without further purification
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 4-trifluoromethyl-benzoic acid methyl ester (3.0 g, 14.7 mmol, 1.0 eq), NaH (1.2 g, 29.4 mmol, 2.0 eq) and propionitrile (4.9 mL, 69.4 mmol, 4.7 eq).
  • the crude product was extracted with DCM and used in the following step without further purification (3.2 g, yield: 96%).
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was washed with basic water and dried, and the title product (2.8 g, yield 84%) was used without further purification
  • the product was prepared according to the general procedure for aminopyrazole synthesis from 4-cyclopropylmethoxy-2-methyl-benzoic acid methyl ester (route A1bis). 0.54 g of the title product was extracted from water and dried (yield 69%) and used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column with gradient elution from 100% EtOAc to EtOAc-MeOH 90:10.
  • the title product (206 mg, yield 36%) was obtained.
  • the product was prepared according to the general procedure from 3-Chloro-4-cyclopropylmethoxy-benzoic acid methyl ester (route A1bis). 0.74 g of the title product was extracted from water and dry (yield 81%) and used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column (gradient elution from 100% EtOAc to EtOAc-MeOH 90:10). 521 mg of the title product (yield 67%) were obtained.
  • the product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl IM and the organic phase separated and dried, to give 1.2 g of the title product (yield 94%) which was used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column (gradient elution from Ethyl Acetate-cycloexane 1:1 to Ethyl Acetate-MeOH 90:10). 650 mg of the title product (yield 52%) were obtained.
  • the product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl IM and the organic phase separated and dried, to give 0.79 g of the title product (yield 79%) which was used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column (gradient elution from EtOAc-cycloexane 1:1 to EtOAc:MeOH 90:10). 810 mg of the title product (yield 97%) were obtained.
  • the product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl 1M and the organic phase separated and dried, to give 0.91 g of the title product (yield 90%) which was used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column (gradient elution from EtOAc-cycloexane 1:1 to Ethyl Acetate:MeOH 90:10). 750 mg of the title product (yield 79%) were obtained.
  • the product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl IM and the organic phase separated and dried, to give 1.24 g of the title product (yield 99%) which was used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through SiO 2 column (gradient elution from EtOAc-cycloexane 1:1 to Ethyl Acetate:MeOH 90:10). 220 mg of the title product (yield 50%) were obtained.
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from pyridine-2-carboxylic acid methyl ester (3.0 g, 21.9 mmol, 1.0 eq). The crude was precipitated from HCl to give the title product as a solid (2.2 g, yield: 69%) which was used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was dissolved in EtOAc, washed with NaHCO 3 , dried and evaporated. NMR analysis showed that a major portion of the crude mixture was still in the opened form: the mixture was then dissolved in CH 3 COOH and heated at 80° C. overnight, to allow for ring closure of the opened form.
  • the product was then recovered as the acylated form, which was de-acylated stirring with HCl 6N at 60° C. overnight obtaining the title product (0.816 g, yield 60%).
  • the product was prepared according to the general procedure for aminopyrazole synthesis (route A1 bis) from 3-difluoromethoxy-benzoic acid methyl ester (1.5 g, 7.4 mmol, 1.0 eq). The crude was precipitated by addition of aqueous HCl to give the product which was used directly for the next step.
  • the product was prepared according to general procedure for aminopyrazole synthesis (route A2).
  • the crude product was purified through Si-column with gradient elution from 100% EtOAc to EtOAc-MeOH 90:10. 1.45 g of title product (yield 87%) was obtained.
  • Bromopyrazole is mixed with K 2 CO 3 and KI in 10 volumes of acetone at room temperature and N-acetylhomopiperazine was added over 1 hr. The reaction mixture was stirred until the reaction was complete. The mixture was filtered, removing the inorganics, washed with acetone and distilled to 2 volumes. The freebase was extracted into methyl THF/EtOH and washed with NaCl and NaHCO 3 . The solvent was replaced with EtOH, a strength of the solution was determined, and 0.93 eq of HCl based on the available freebase was added to a mixture of acetone, ethanol and water. Careful monitoring of the pH yielded crystalline product in a 70% overall yield and the desired form 1.
  • the 2-methoxy derivative was purified by SiO 2 chromatography, eluting with a DCM/MeOH gradient (from 100% DCM to 90/10 DCM/MeOH); the 3-methoxy derivative was triturated with Et 2 O. Yields were generally 65-90%.
  • N-acetylhomopiperazine (0.062 kg, 0.057 L, 0.434 mol) was added via addition funnel to the reactor over a minimum of 45 min., maintaining the temperature in the range of 25-30° C.
  • the addition funnel was rinsed with 0.05 L acetone.
  • a white mixture persisted.
  • the mixture was stirred (235 rpm) in the range of 25-30° C. for a minimum of 16 h, forming a white/yellow mixture.
  • the reaction progress was monitored by HPLC and was considered complete when there was ⁇ 2% of the starting material (bromopyrazole) and ⁇ 2% of the iodopyrazole present.
  • the reactor contents were cooled to 5-15° C. over a minimum of 15 min with agitation (295 rpm) to form a white/yellow mixture that was then stirred for a minimum of 1 h.
  • the mixture was then filtered on a Buchner funnel with filter paper using house vacuum for 1.5 min.
  • the cake was washed twice with acetone (total of 0.24 kg, 0.30 L) at 5-15° C.
  • the wash was combined with the mother liquor from the prior filtration and used to rinse the reactor.
  • the filtrate was concentrated to a volume of approximately 0.45 L to form a clear solution.
  • the organic layer was retained. A homogeneous 5% solution of sodium bicarbonate (0.03 kg) in water (0.57 L) at 25° C. was used to wash organic layer, stirring for a minimum of 5 min. at 10-15° C. The agitation was stopped to allow the layers to settle, the product being in the upper layer. The layers were separated, keeping any emulsion in the upper organic layer. The organic layer was retained and concentrated to a volume of 0.35 L, forming a hazy solution. The mixture was chased with ethanol to remove residual water.
  • a solution of 5% HCl (0.042 kg, 0.036 L) in acetone (0.174 L) and alcohol solution (0.0174 L of ethanol:acetone (91:9) v/v) was prepared and stirred until homogeneous at 10° C.
  • 0.05 L of water was added to form a clear solution.
  • One third of the 5% HCl solution (0.076 L) was added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C.
  • a second third of the 5% HCl solution (0.076 L) was then added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C.
  • the contents of the reactor were seeded with 75 mg of 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide HCl (e.g., Form 1), followed by the addition of the last third of 5% HCl solution (0.076 L) over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. Another 0.08 equiv. of the 5% HCl solution (0.023 L) was then added to the reactor over a minimum of 30 min., maintaining the temperature in the range of 20-25° C. Judicious monitoring of pH was performed to attain the desired pH range of 5.2-5.8. Based on the strength calculation, 0.85 equiv. of acid was added over 1 hr and the remaining acid was added over a minimum of 30 min. with careful monitoring of pH.
  • 5% HCl solution 0.076 L
  • the mixture was stirred at 20-25° C. for a minimum of 1 hr., forming a thin suspension.
  • Acetone (0.6 L) was added over a minimum of 60 min., maintaining the temperature in the range of 20-25° C.
  • the mixture was stirred at 20-25° C. for a minimum of 60 min.
  • Acetone (1.5 L) was added to the reactor over a minimum of 3 hr., maintaining the temperature in the range of 20-25° C., forming a thick suspension.
  • the mixture was then stirred at 20-25° C. for a minimum of 12 h. Crystallization was considered complete when there was ⁇ 20% of the product present in the mother liquor. Longer stirring was employed if crystallization was not complete.
  • the present Example describes the preparation of the hydrochloride salt form of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide.
  • the hydrochloric acid salt form readily adopted a solid form. Indeed, at least four different crystalline forms (i.e., polymorphs) were observed for the hydrochloric acid salt form (see below).
  • Differential scanning calorimetry data were collected for each solid form achieved using a DSC (TA instruments, model Q1000) under the following parameters: 50 mL/min purge gas (N2); scan range 40 to 200° C., scan rate 10° C./min.
  • Thermogravimetric analysis data were collected using a TGA instruments (Mettler Toledo, model TGA/SDTA 851e) under the following parameters: 40 ml/min purge gas (N2); scan range 30 to 250° C., scan rate 10° C./min.
  • X-ray data were acquired using an X-ray powder diffractometer (Bruker-axs, model D8 advance) having the following parameters: voltage 40 kV, current 40.0 mA, scan range 5 to 30°, scan step size 0.01°, total scan time 33 minutes, VANTEC detector, and antiscattering slit 1 mm.
  • FIGS. 1-7 show characterization data for hydrochloride salt forms.
  • the hydrochloride salt was polymorphic, adopting crystalline forms exhibiting DSC endotherms at 119° C. (Form III), 127° C. (Form IV), 167° C. (Form II), and 186° C. (Form I).
  • the Table below illustrates certain characteristics of observed hydrochloride salt crystal forms:
  • Form I (186° C.) is relatively non-hygroscopic, gaining only about 0.5% moisture when equilibrated at RH less than or equal to 70%. At 70-100% RH, Form I gains at least about 2% moisture, but loses it without significant hysteresis on decreasing RH. Evidence of a hydrochloride hydrate was not observed after the hygroscopicity test.
  • the present Example describes characterization of two surprisingly non-hygroscopic crystal forms (Forms I and II, as described above) of a hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide:
  • Form I is 185° C. (plus or minus 2 degrees); the melting point of Form II is 166° C. (plus or minus 5 degrees).
  • Form I picks up moisture at relative humidity (RH) of about 50% and absorbs up to about 2% water eventually (90% RH) and loses the water as RH decreases ( ⁇ 50%).
  • RH relative humidity
  • Form I also exhibits characteristic X-ray peaks at 20 of 15.3° and 21.9°, plus or minus about 0.3°, depending upon the machine and measurement method utilized.
  • Form II picks up moisture at RH of about 20% and absorbs up to 7% water eventually (RH of 90%) and holds 2% at low RH (0%).
  • Form II also exhibits characteristic X-ray peaks at 2 ⁇ of 20.2° and 24.9°, plus or minus about 0.3°, depending upon the machine and measurement method utilized.
  • Differential scanning calorimetry data were collected for each solid form achieved using a DSC (TA instruments, model Q1000) under the following parameters: 50 mL/min purge gas (N2); scan range 40 to 200° C., scan rate 10° C./min.
  • Thermogravimetric analysis data were collected using a TGA instruments (Mettler Toledo, model TGA/SDTA 851e) under the following parameters: 40 ml/min purge gas (N2); scan range 30 to 250° C., scan rate 10° C./min.
  • X-ray data were acquired using an X-ray powder diffractometer (Bruker-axs, model D8 advance) having the following parameters: voltage 40 kV, current 40.0 mA, scan range (2 ⁇ ) 3.7 to 30°, scan step size 0.0°, total scan time 33 minutes, VANTEC detector, and antiscattering slit 1 mm.
  • the present Example describes the preparation of crystal form I of the hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide.
  • a seed of crystal form I of the hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide was added. 0.18 ml diluted HCl solution was added slowly. Around two minutes later, 0.18 ml diluted HCl solution was added slowly. Around two minutes later, another 0.18 ml diluted HCl solution was added slowly. The mixture was heated to about 40-50° C., and then was left at room temperature while stirring overnight. The crystals were filtered and washed with 1.5 ml acetone, and were dried at 45° C. for about 6 hours.
  • the hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide may be re-purified by basifying the hydrochloride salt and extracting the free base into a suitable solvent (e.g. methylene chloride). The organic extracts may be washed with water. The organic phase is concentrated and the solvent switched to ethanol. Acetone is added to give a solution of compound A which was clarified and mixed with ethanol, acetone, hydrochloric acid, and water. Acetone is added, the solids filtered, washed with mixture of acetone, water and dried to give compound I. A representative procedure is described below.
  • Acetone (0.63 kg) was added to the concentrate and the solution clarified. An accurate strength of the free base was determined of the concentrate. Water (0.065 kg) was added to form a clear solution. A solution of 5% HCl (0.043 kg) in acetone (0.14 kg) and alcohol (0.14 kg of ethanol:acetone (91:9) v/v) was prepared and stirred until homogeneous at 10° C. About one third of the 5% HCl solution (0.098 kg) was added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. A second third of the 5% HCl solution (0.098 kg) was then added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C.
  • the contents of the reactor were seeded with 75 mg of 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide HCl (e.g., Form 1), followed by the addition of the last third of 5% HCl solution (0.098 kg) over a minimum of 20 min., maintaining the temperature in the range of 20-25° C.
  • HCl e.g., Form 1
  • the contents of the reactor were seeded with another 75 mg of 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide HCl (e.g., Form 1).
  • Another 0.08 equiv. of the 5% HCl solution (0.029 kg) was then added to the reactor over a minimum of 30 min., maintaining the temperature in the range of 20-25° C.
  • Judicious monitoring of pH was performed to attain the desired pH range of 5.2-5.8.
  • the mixture was stirred at 20-25° C. for a minimum of 1 hr., forming a thin suspension.
  • Acetone (0.63 kg) was added over a minimum of 60 min., maintaining the temperature in the range of 20-25° C.
  • the mixture was stirred at 20-25° C. for a minimum of 60 min.
  • Acetone (1.58 kg) was added to the reactor over a minimum of 3 hr., maintaining the temperature in the range of 20-25° C., forming a thick suspension.
  • the mixture was then stirred at 20-25° C. for a minimum of 12 h. Crystallization was considered complete when there was ⁇ 15% of the product present in the mother liquor. Longer stirring was employed if crystallization was not complete.
  • the mixture was then filtered on a Buchner funnel (polypropylene pad) using house vacuum.
  • a solution of water (0.012 kg), acetone (0.24 kg) and 0.063 kg alcohol (ethanol:acetone (91:9) v/v) was stirred until homogeneous (20% ethanol, 3% water, 77% acetone overall). This solution was used to wash the filter cake.
  • a solution of water (0.012 kg), acetone (0.18 kg) and 0.13 kg alcohol (ethanol:acetone (91:9) v/v) was stirred until homogeneous (40% ethanol, 3% water, 57% acetone overall). This solution was used to wash the filter cake.
  • the wet cake was subjected to suction under nitrogen using house vacuum and held for 30 min. after dripping stopped.

Abstract

The present invention provides compounds and compositions, methods of making them, and methods of using them to modulate α7 nicotinic acetylcholine receptors and/or to treat any of a variety of disorders, diseases, and conditions. Provided compounds can affect, among other things, neurological, psychiatric and/or inflammatory system.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. provisional patent application Ser. No. 61/021,015, filed Jan. 14, 2008, the entirety of which is hereby incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to the synthesis of compounds with α7 nicotinic acetylcholine receptor (α7 nAChR) agonistic activity, derivatives thereof, and intermediates thereto.
  • BACKGROUND OF THE INVENTION
  • Agents that bind to nicotinic acetylcholine receptors have been indicated as useful in the treatment and/or prophylaxis of various diseases and conditions, particularly psychotic diseases, neurodegenerative diseases involving a dysfunction of the cholinergic system, and conditions of memory and/or cognition impairment, including for example, schizophrenia, anxiety, mania, depression, manic depression, Tourette's syndrome, Parkinson's disease, Huntington's disease, cognitive disorders (such as Alzheimer's disease, Lewy Body Dementia, Amyotrophic Lateral Sclerosis, memory impairment, memory loss, cognition deficit, attention deficit, Attention Deficit Hyperactivity Disorder), and other uses such as treatment of nicotine addiction, inducing smoking cessation, treating pain (e.g. analgesic use), providing neuroprotection, and treating jetlag. See for example WO 97/30998; WO 99/03850; WO 00/42044; WO 01/36417; Holladay et al., J. Med. Chem., 40:26, 4169-94 (1997); Schmitt et al., Annual Reports Med. Chem., Chapter 5, 41-51 (2000); Stevens et al., Psychopharmatology, (1998) 136: 320-27; and Shytle et al., Molecular Psychiatry, (2002), 7, pp. 525-535.
  • Different heterocyclic compounds carrying a basic nitrogen and exhibiting nicotinic and muscarinic acetylcholine receptor affinity or claimed for use in Alzheimer's disease have been described, e.g. 1H-pyrazole and pyrrole-azabicyclic compounds (WO2004013137); nicotinic acetylcholine agonists (WO2004039366); ureido-pyrazole derivatives (WO0112188); oxadiazole derivatives having acetylcholinesterase-inhibitory activity and muscarinic agonist activity (WO9313083); pyrazole-3-carboxylic acid amide derivatives as pharmaceutical compounds (WO2006077428); arylpiperidines (WO2004006924); ureidoalkylpiperidines (U.S. Pat. No. 6,605,623); compounds with activity on muscarinic receptors (WO9950247). In addition, modulators of alpha7 nicotinic acetylcholine receptor are disclosed in WO06008133, in the name of the same applicant.
  • SUMMARY
  • Among other things, the invention provides methods of preparing compounds acting as full or partial agonists at the α7 nicotinic acetylcholine receptor (α7 nAChR), and intermediates thereof. Such compounds are useful for the treatment of diseases that may benefit from the activation of the alpha 7 nicotinic acetylcholine receptor such as neurological, neurodegenerative, psychiatric, cognitive, immunological, inflammatory, metabolic, addiction, nociceptive, and sexual disorders, in particular Alzheimer's disease, schizophrenia, and/or others. Such compounds include those of formula I-1:
  • Figure US20090264648A1-20091022-C00001
  • or a pharmaceutically acceptable salt thereof, wherein each of Ar, Y, Y′, T, and Ring A is as defined herein.
  • The present invention also provides synthetic intermediates useful for preparing such compounds. The invention further provides methods to provide cost effective yields and purity.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIGS. 1-7 show characterization data for hydrochloride salts.
  • FIG. 8 illustrates the effect of pH and HCl equivalence on HCl salt formation.
  • FIG. 9 shows the effects of pH and HCl equivalence on HCl salt formation
  • FIGS. 10 and 11 depict conversion of higher HCl salts to mono-HCl forms.
  • FIG. 12 shows a DSC scan of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIG. 13 shows a TGA thermogram of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIGS. 14 a-b show X-ray diffraction pattern and data for 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIG. 15 presents DVS isothermal analysis of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form I.
  • FIG. 16 is a DSC scan of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • FIG. 17 is a TGA thermogram of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • FIGS. 18 a-b show X-ray diffraction pattern and data for 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • FIG. 19 presents a DVS isothermal analysis of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloric salt Form II.
  • DESCRIPTION OF CERTAIN PARTICULAR EMBODIMENTS Definitions
  • The term “aliphatic” or “aliphatic group,” as used herein, means a straight-chain (i.e., unbranched) or branched, hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocycle” “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule. In certain embodiments, aliphatic groups contain 1-6 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms. In some embodiments, “cycloaliphatic” (or “carbocycle”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Such cycloaliphatic groups include cycloalkyl and cycloalkenyl groups. Suitable aliphatic groups include, but are not limited to, linear or branched alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • The term “lower alkyl,” as used herein, refers to a hydrocarbon chain having up to 6 carbon atoms, preferably 1 to 3 carbon atoms, and more preferably 1 to 2 carbon atoms. The term “alkyl” includes, but is not limited to, straight and branched chains such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, or t-butyl.
  • The term “alkoxy,” as used herein, refers to the group —OR*, wherein R* is a lower alkyl group.
  • The term “acyl,” as used herein, refers to a group having the general formula —C(═O)RX1, —C(═O)ORX1, —C(═O)—O—C(═O)RX1, —C(═O)SRX1, —C(═O)N(RX1)2, —C(═S)RX1, —C(═S)N(RX1)2, and —C(═S)S(RX1), —C(═NRX1)RX1, —C(═NRXI)ORXI, —C(═NRXI)SRX1, and —C(═NRX1)N(RX1)2, wherein RXI is hydrogen; halogen; substituted or unsubstituted hydroxyl; substituted or unsubstituted thiol; substituted or unsubstituted amino; substituted or unsubstituted acyl, cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched alkyl; cyclic or acyclic, substituted or unsubstituted, branched or unbranched alkenyl; substituted or unsubstituted alkynyl; substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy, mono- or di-aliphaticamino, mono- or di-heteroaliphaticamino, mono- or di-alkylamino, mono- or di-heteroalkylamino, mono- or di-arylamino, or mono- or di-heteroarylamino; or two RXI groups taken together form a 5- to 6-membered heterocyclic ring. Exemplary acyl groups include aldehydes (—CHO), carboxylic acids (—CO2H), ketones, acyl halides, esters, amides, imines, carbonates, carbamates, and ureas. Acyl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy, acyloxy, and the like, each of which may or may not be further substituted).
  • The terms “halogen” or “halo,” as used herein, refer to chlorine, bromine, fluorine or iodine.
  • The term “alkenyl,” as used herein refers to an aliphatic straight or branched hydrocarbon chain having 2 to 4 carbon atoms that has one or more double bonds. Examples of alkenyl groups include vinyl, prop-1-enyl, allyl, methallyl, but-1-enyl, but-2-enyl, or but-3-enyl. The term “lower alkenyl” refers to an alkenyl group having 1 to 3 carbon atoms.
  • The term “aryl,” as used herein refers to phenyl or an 8-10 membered bicyclic partially unsaturated or aryl ring. Exemplary aryl groups include phenyl and naphthyl. In certain embodiments, the term “aryl,” as used herein refers to an 8-10 membered bicyclic partially unsaturated the wherein at least one of the rings is aromatic.
  • The term “heteroaryl,” as used herein, refers to a 5-6 membered monocyclic heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic partially unsaturated or heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Examples of heteroaryls include, but are not limited to, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, indazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, isobenzothienyl, quinolyl, isoquinolyl, quinoxalinyl, or quinazolinyl.
  • As described herein, compounds of the invention may contain “optionally substituted” moieties. In general, the term “substituted,” whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term “stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group are independently halogen; —(CH2)0-4R; —(CH2)0-4OR; —O(CH2)0-4R, —O—(CH2)0-4C(O)OR; —(CH2)0-4CH(OR)2; —(CH2)0-4SR; —(CH2)0-4Ph, which may be substituted with R; —(CH2)0-4O(CH2)0-1Ph which may be substituted with R; —CH═CHPh, which may be substituted with R; —(CH2)0-4O(CH2)0-1-pyridyl which may be substituted with R; —NO2; —CN; —N3; —(CH2)0-4N(R)2; —(CH2)0-4N(R)C(O)R; —N(R)C(S)R; —(CH2)0-4N(R)C(O)NR 2; —N(R)C(S)NR 2; —(CH2)0-4N(R)C(O)OR; —N(R)N(R)C(O)R; —N(R)N(R)C(O)NR 2; —N(R)N(R)C(O)OR; —(CH2)0-4C(O)R; —C(S)R; —(CH2)0-4C(O)OR; —(CH2)0-4C(O)SR; —(CH2)0-4C(O)OSiR 3; —(CH2)0-4OC(O)R; —OC(O)(CH2)0-4SR—, SC(S)SR; —(CH2)0-4SC(O)R; —(CH2)0-4C(O)NR 2; —C(S)NR 2; —C(S)SR; —SC(S)SR, —(CH2)0-4OC(O)NR 2; —C(O)N(OR)R; —C(O)C(O)R; —C(O)CH2C(O)R; —C(NOR)R; —(CH2)0-4SSR; —(CH2)0-4S(O)2R; —(CH2)0-4S(O)2OR; —(CH2)0-4OS(O)2R; —S(O)2NR 2; —(CH2)0-4S(O)R; —N(R)S(O)2NR 2; —N(R)S(O)2R; —N(OR)R; —C(NH)NR 2; —P(O)2R; —P(O)R 2; —OP(O)R 2; —OP(O)(OR)2; SiR 3; —(C1-4 straight or branched alkylene)O—N(R)2; or —(C1-4 straight or branched alkylene)C(O)O—N(R)2, wherein each R may be substituted as defined below and is independently hydrogen, C1-6 aliphatic, —CH2Ph, —O(CH2)0-1Ph, —CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.
  • Suitable monovalent substituents on R (or the ring formed by taking two independent occurrences of R together with their intervening atoms), are independently halogen, —(CH2)0-2R, -(haloR), —(CH2)0-2OH, —(CH2)0-2OR, —(CH2)0-2CH(OR)2; —O(haloR), —CN, —N3, —(CH2)0-2C(O)R, —(CH2)0-2C(O)OH, —(CH2)0-2C(O)OR, —(CH2)0-2SR, —(CH2)0-2SH, —(CH2)0-2NH2, —(CH2)0-2NHR, —(CH2)0-2NR 2, —NO2, —SiR 3, —OSiR 3, —C(O)SR, —(C1-4 straight or branched alkylene)C(O)OR, or —SSR wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently selected from C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R include ═O and ═S.
  • Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: ═O, ═S, ═NNR*2, ═NNHC(O)R*, ═NNHC(O)OR*, ═NNHS(O)2R*, ═NR*, ═NOR*, —(C(R*2))2-3O—, or S(C(R*2))2-3S—, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: —O(CR*2)2-3O—, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R* include halogen, —R, -(haloR), —OH, —ORØ, —O(haloR), —CN, —C(O)OH, —C(O)OR, —NH2, —NHR, —NR 2, or —NO2, wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include —R, —NR 2, —C(O)R, —C(O)OR, —C(O)C(O)R, —C(O)CH2C(O)R, —S(O)2R, —S(O)2NR 2, —C(S)NR 2, —C(NH)NR 2, or —N(R)S(O)2R; wherein each R is independently hydrogen, C1-6 aliphatic which may be substituted as defined below, unsubstituted —OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R are independently halogen, —R, -(haloR), —OH, —OR, —O(haloR), —CN, —C(O)OH, —C(O)OR, —NH2, —NHR, —NR 2, or —NO2, wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • The term “pharmaceutically acceptable salts” or “pharmaceutically acceptable salt” includes acid addition salts, that is salts derived from treating compounds of formulae I-1 and A with an organic or inorganic acid such as, for example, acetic, lactic, citric, cinnamic, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, oxalic, propionic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, glycolic, pyruvic, methanesulfonic, ethanesulfonic, toluenesulfonic, salicylic, benzoic, or similarly known acceptable acids. Where a compound of formulae I-1 and A contains a substituent with acidic properties, for instance, phenolic hydroxyl, —SO2H or —CO2H, the term also includes salts derived from bases, for example, sodium salts.
  • In certain embodiments, the present invention provides a method for preparing compounds of formula I-1:
  • Figure US20090264648A1-20091022-C00002
  • or a pharmaceutically acceptable salt thereof, wherein:
    • Ring A is a 4-8 membered saturated ring, having 0-2 heteroatoms independently selected from O, N, or S in addition to the nitrogen depicted in Ring A, wherein Ring A is independently substituted with 0-4 R′ groups
    • R′ is selected from the group consisting of mono- or di-[linear, branched or cyclic C1-6 alkyl]aminocarbonyl; linear, branched or cyclic C1-6 alkyl, alkoxy, or acyl;
    • Y and Y′ are each independently N or C, with the proviso that at least one of Y or Y′ is N;
    • T is a C3-5 bivalent hydrocarbon chain, optionally carrying an oxo group and optionally substituted with one or more halogen, hydroxy, C1-5 alkyl, alkoxy, fluoroalkyl, hydroxyalkyl, alkylidene, or fluoroalkylidene groups; C3-6 cycloalkane-1,1-diyl, oxacycloalkane-1,1-diyl, C3-6 cycloalkane-1,2-diyl, or oxacycloalkane-1,2-diyl groups, wherein the bonds of the 1,2-diyl radical form a fused ring with the T chain; and with the proviso that when T carries an oxo group, said oxo group is not part of an amide bond; and
    • Ar is a group selected from 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur; wherein Ar is optionally substituted with one or more substituents independently selected from halogen; hydroxy; mercapto; cyano; nitro; amino; sulfonyl; linear, branched or cyclic (C1-C6) alkyl, trihaloalkyl, di- or trihaloalkoxy, alkoxy, or alkylcarbonyl; (C3-C6) cycloalkyl-(C1-C6) alkoxy; (C3-C6) cycloalkyl-(C1-C6) alkyl; linear, branched, or cyclic (C1-C6) alkylcarbonylamino; mono- or di-, linear, branched, or cyclic (C1-C6) alkylaminocarbonyl; carbamoyl; linear, branched, or cyclic (C1-C6) alkylsulphonylamino; linear, branched, or cyclic (C1-C6) alkylsulphonyl; mono- or di-, linear, branched, or cyclic (C1-C6) alkylsulphamoyl; linear, branched or cyclic (C1-C6) alkoxy-(C1-C6) alkyl; wherein, two substituents may be taken together with their intervening atoms to form a ring.
  • In certain embodiments, the present compounds are generally prepared according to Scheme 1 set forth below:
  • Figure US20090264648A1-20091022-C00003
  • wherein each Ring A, Y, Y′, T, and Ar is defined in formula I-1 and described in classes and subclasses above and herein, and LG2 and LG3 are leaving groups.
  • In certain embodiments, the T moiety on formulae I-1, B′, and F′ is a C3-5 bivalent hydrocarbon chain, optionally substituted with one or more halogen, hydroxy, C1-5 alkyl, alkoxy, fluoroalkyl, hydroxyalkyl, alkylidene, or fluoroalkylidene groups. In some embodiments, the T moiety is a C4 bilvalent hydrocarbon chain.
  • In certain embodiments, Y is a nitrogen atom. In certain embodiments, Y′ is a nitrogen atom.
  • In certain embodiments, Ring A of a compound of formula G′ is a 4-8 membered saturated ring. In certain embodiments, a compound of formula G′ is a piperazine derivative. In certain embodiments, a compound of formula G′ is N-acetylhomopiperazine.
  • In certain embodiments, Ar is an optionally substituted group selected from 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. In some embodiments, Ar is an optionally substituted 6 membered aryl group. In certain embodiments, Ar is methoxy-phenyl. In certain embodiments, Ar is 4-methoxy-phenyl. In some embodiments, Ar is hydroxy-phenyl. In some embodiments, Ar is a phenyl group substituted with a —OSO3H group.
  • At step S-1, a compound of formula C′ is reacted in a suitable solvent, as defined and described herein, with a compound of formula F′ to form a compound of formula B′.
  • The LG2 group of formulae B′ and F′ is a suitable leaving group, as defined and described herein. In certain embodiments, the LG2 group of formulae B′ and F′ is halogen, —OMs, —OTs, or —OTf. In certain embodiments, the LG2 group of formulae B′ and F′ is —Br.
  • The LG3 group of formula F′ is a suitable leaving group, as defined and described herein. In certain embodiments, the LG3 group of formula F′ is halogen, —OR,
  • Figure US20090264648A1-20091022-C00004
  • wherein each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. Examples of LG3 groups of formula F′ include —OH, —OMe, —OEt, —Cl, —Br,
  • Figure US20090264648A1-20091022-C00005
  • and the like. In some embodiments, LG3 is —Cl.
  • In certain embodiments, LG3 is —OH, and the reaction between a compound of formula F′ and a compound of formula B′ is carried out using suitable peptide coupling conditions. Suitable peptide coupling conditions are well known in the art and include those described in detail in Han et al., Tetrahedron, 60, 2447-67 (2004), the entirety of which is hereby incorporated by reference. In certain embodiments, the peptide coupling conditions include the addition of HOBt, DMAP, BOP, HBTU, HATU, BOMI, DCC, EDC, IBCF, or a combination thereof.
  • In certain embodiments, the compound of formula F′ is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F′ is 5-bromovaleryl chloride.
  • One of ordinary skill in the art will appreciate that a variety of suitable leaving groups LG3 can be used to facilitate the reaction described in step S-1, and all such suitable leaving groups are contemplated by the present invention.
  • Suitable solvents for step S-1 include aprotic solvents, aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, aliphatic ethers or combinations thereof. In certain embodiments, the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof. In some embodiments, the solvent is a mixture of acetonitrile and DMF. In certain embodiments, the solvent is a 9:1 mixture of acetonitrile:DMF.
  • Suitable bases for step S-1 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof. In certain embodiments, the base is diisopropylethylamine.
  • Suitable temperatures at which the reaction described in step S-1 may occur include about −20° C. to about 60° C. In certain embodiments, the temperature is about −10° C. to about 25° C. In some embodiments, the temperature is about −10° C.
  • While not wishing to be bound by any particular theory, it is believed that the order of reagent addition may be useful in reducing the formation of byproducts in step S-1. In certain embodiments a compound of formula F′ is added slowly to a compound of formula C′.
  • It will be appreciated that certain reaction conditions may result in the formation of a regioisomer in step S-1 (i.e., reaction of endocyclic pyrazole nitrogen with a compound of formula F′). In certain embodiments, the product of step S-1 is treated to remove the undesired regioisomer. In some embodiments, the product of step S-1 is re-slurried with a particular solvent or solvents to remove the regioisomer. In some embodiments, the product of step S-1 is re-slurried with t-butyl methyl ether to remove the regioisomer.
  • At step S-2, a compound of formula B′ is reacted in a suitable solvent with a compound of formula G′, optionally in the presence of a suitable base and/or cataylst, to produce compound I-1. One of ordinary skill in the art will recognize that when a compound of formula B′ is 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom. Examples of such iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof. While not wishing to be bound by any particular theory, it is believed that 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be a more reactive species in step S-2 than 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, due to the greater leaving group character of iodide over bromide.
  • Suitable solvents for step S-2 include aprotic solvents, aliphatic halides, aliphatic ethers or combinations thereof. In certain embodiments, the solvent is selected from dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof. In some embodiments, the solvent is acetone.
  • Suitable bases for step S-2 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof. In certain embodiments, the base is potassium carbonate. In certain embodiments, the base is diisopropylethylamine.
  • In certain embodiments, the iodide source used in step S-2 is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B′. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B′.
  • Suitable temperatures at which the reaction described in step S-2 may occur include about −20° C. to about 60° C. In certain embodiments, the temperature is about −10° C. to about 25° C. In some embodiments, the temperature is about 25° C.
  • One of ordinary skill in the art will recognize that a compound of formula I-1 may be further transformed into a pharmaceutically acceptable salt. In certain embodiments, the pharmaceutically acceptable salt is a hydrochloride salt. In some embodiments, the pharmaceutically acceptable salt is a mono hydrochloride salt.
  • In certain embodiments, as described in Example 5, an extractive workup step may be performed following step S-2. In certain embodiments, the organic phase comprises one or more organic solvents. In some embodiments, the organic solvents are ethanol, methyl tetrahydrofuran, dichloromethane, or a combination thereof. In certain embodiments, the solvents are ethanol and methyl tetrahydrofuran. In certain embodiments, the solvents are ethanol and dichloromethane. In some embodiments, the organic solvents are 5% ethanol in methyl tetrahydrofuran. In some embodiments, the organic solvents are 5% ethanol in dichloromethane.
  • In certain embodiments, dimer compounds may be formed as side products at step S-2. In such cases, it has been found that performing additional washes following HCl salt formation may be useful for reducing the presence of such dimer products.
  • In certain embodiments, each of the aforementioned synthetic steps may be performed sequentially with isolation of each intermediate B′ and I-1 performed after each step. Alternatively, each of steps S-1 and S-2, as depicted in Scheme I above, as well as any subsequent salt formation, may be performed in a manner whereby no isolation of one or more intermediates B′ and I-1 is performed. In certain embodiments, steps S-1 and S-2 are performed in sequence without any isolation of intermediates. In certain embodiments, step S-2 and salt formation are performed without any isolation of intermediates. While not wishing to be bound by any particular theory, it is believed that such techniques may be useful in obtaining compounds of formulae B′, I-1, and pharmaceutically acceptable salts thereof in greater yield and purity.
  • In certain embodiments, the present invention provides a method for preparing compound 1,5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrogen chloride:
  • Figure US20090264648A1-20091022-C00006
  • which may also be referred to as 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide.
  • Compound I, a potent α7 nAChR agonist, is effective in treating diseases that may benefit from the activation of the alpha 7 nicotinic acetylcholine receptor such as neurological, neurodegenerative, psychiatric, cognitive, immunological, inflammatory, metabolic, addiction, nociceptive, and sexual disorders, in particular Alzheimer's disease, schizophrenia, and/or others.
  • Certain methods of preparing compounds of the present invention are known in the art and include those described in detail in WO2006/008133 and U.S. Ser. No. 60/880,629, the entirety of each of which is hereby incorporated herein by reference.
  • In certain embodiments, the present compounds are generally prepared according to Scheme 2 set forth below:
  • Figure US20090264648A1-20091022-C00007
  • In one aspect, the present invention provides methods for preparing a free base, A, according to the steps depicted in Scheme 2, above. At step S-3, an aromatic ester of formula E is reacted, optionally in a suitable solvent, with a suitable base and acetonitrile to provide β-ketonitrile D. Such suitable bases include metal alkoxides and metal hydrides. In certain embodiments, the base is an alkali hydride, a metal alkyl, a metal amide, or a metal silazide. Other suitable bases include KH, n-butyl lithium, hexyl lithium, lithium diisopropylamide, Li—N(Si-alkyl)2, lithium hexamethyldisilazide (LiHMDS), sodium hexamethyldisilazide (NaHMDS), potassium hexamethyldisilazide (KHMDS) potassium t-amylate, sodium t-butoxide (NaOtBu), and sodium hydride (NaH). In certain embodiments, the base is LiHMDS.
  • The acetonitrile used in step S-3 may be used in a range of equivalents relative to aromatic ester E. In certain embodiments, the equivalents of acetonitrile range from 0.5 to 50 equivalents. In certain embodiments, the equivalents of acetonitrile range from 2 to 10 equivalents. In some embodiments, the equivalents of acetonitrile range from 4 to 6 equivalents.
  • Suitable temperatures at which the reaction described in step S-3 may occur include about −20° C. to about 80° C. In certain embodiments, the temperature is about −20° C. to about 0° C. In some embodiments, the temperature is about −10° C.
  • Step S-3 may optionally employ a suitable solvent. A suitable solvent is a solvent or a solvent mixture that, in combination with the combined reacting partners and reagents, facilitates the progress and/or rate of the reaction. The suitable solvent may solubilize one or more of the reaction components, or, alternatively, the suitable solvent may facilitate the suspension of one or more of the reaction components; see, generally, “Advanced Organic Chemistry,” Jerry March, 5th edition, John Wiley and Sons, N.Y.
  • Examples of solvents suitable for use in step S-3 include anhydrous aprotic solvents, such as aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, aliphatic nitriles, and aliphatic ethers. In some embodiments, the solvent is selected from toluene, acetonitrile, diethyl ether, t-butyl methyl ether, THF, benzene, dichloromethane or combinations thereof. In certain embodiments, no solvent is used.
  • The LG1 group of formula E is a suitable leaving group. A suitable leaving group is a chemical group that is readily displaced by a desired incoming chemical moiety. Suitable leaving groups are well known in the art, e.g., see, “Advanced Organic Chemistry,” Jerry March, 5th Ed., pp. 351-357, John Wiley and Sons, N.Y. Such leaving groups include, but are not limited to, halogen, alkoxy, sulphonyloxy, optionally substituted alkylsulphonyl, optionally substituted alkenylsulfonyl, optionally substituted arylsulfonyl, and diazonium moieties. Examples of some suitable leaving groups include chloro, iodo, bromo, fluoro, methanesulfonyl (mesyl), tosyl, triflate, nitro-phenylsulfonyl (nosyl), and bromo-phenylsulfonyl (brosyl).
  • In certain embodiments, the LG1 group of formula E is halogen, —OR,
  • Figure US20090264648A1-20091022-C00008
  • wherein each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. Examples of LG1 groups of formula E include —OH, —OMe, —OEt, Cl, Br,
  • Figure US20090264648A1-20091022-C00009
  • and the like. In some embodiments, LG1 is —OR, wherein R is C1-6 alkyl. In certain embodiments, LG1 is —OR, wherein R is methyl.
  • One of ordinary skill in the art will appreciate that a variety of suitable leaving groups can be used to facilitate the reaction described in step S-3, and all such suitable leaving groups are contemplated by the present invention.
  • According to an alternate embodiment, the suitable leaving group may be generated in situ within the reaction medium. For example, a leaving group may be generated in situ from a precursor of that compound wherein said precursor contains a group readily replaced by said leaving group in situ.
  • At step S-4, β-ketonitrile D is reacted in a suitable solvent with hydrazine, or an equivalent thereof, to form aryl aminopyrazole C. Such hydrazine equivalents are well known to one of ordinary skill in the art and include, but are not limited to, anhydrates, hydrates, monohydrates, monohydrochlorides, dihydrochlorides, and sulfates. In certain embodiments, the hydrazine equivalent used in step S-4 is a hydrate. In some embodiments, the hydrazine equivalent is a monohydrate.
  • Suitable solvents for step S-4 include alkyl alcohols, such as C1 to C4 alcohols (e.g. ethanol, methanol, 2-propanol), aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, or aliphatic ethers or combinations thereof. In certain embodiments, the solvent is selected from ethanol, toluene, dichloromethane, diethyl ether, THF, benzene or combination thereof. In some embodiments, the solvent is ethanol.
  • Suitable temperatures at which the reaction described in step S-4 may occur include about 10° C. to about 150° C. In certain embodiments, the temperature is about 30° C. to about 70° C. In some embodiments, the temperature is about 60° C.
  • At step S-5, aryl aminopyrazole C is reacted in a suitable solvent with a compound of formula F to form a compound of formula B.
  • The LG2 group of formulae B and F is a suitable leaving group, as defined and described herein. In certain embodiments, the LG2 group of formulae B and F is halogen, —OMs, —OTs, or —OTf. Examples of the LG2 group of formulae B and F include —Br, —I, —OMs, —OTs, and —OTf.
  • The LG3 group of formula F is a suitable leaving group, as defined and described herein. In certain embodiments, the LG3 group of formula F is halogen, —OR,
  • Figure US20090264648A1-20091022-C00010
  • wherein each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. Examples of LG3 groups of formula F include —OH, —OMe, —OEt, —Cl, —Br,
  • Figure US20090264648A1-20091022-C00011
  • and the like. In some embodiments, LG3 is —Cl.
  • In certain embodiments, LG3 is —OH, and the reaction between a compound of formula F and a compound of formula B is carried out using suitable peptide coupling conditions. Suitable peptide coupling conditions are well known in the art and include those described in detail in Han et al., Tetrahedron, 60, 2447-67 (2004), the entirety of which is hereby incorporated by reference. In certain embodiments, the peptide coupling conditions include the addition of HOBt, DMAP, BOP, HBTU, HATU, BOMI, DCC, EDC, IBCF, or a combination thereof.
  • In certain embodiments, the compound of formula F is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F is 5-bromovaleryl chloride.
  • One of ordinary skill in the art will appreciate that a variety of suitable leaving groups LG3 can be used to facilitate the reaction described in step S-5, and all such suitable leaving groups are contemplated by the present invention.
  • Suitable solvents for step S-5 include aprotic solvents, aliphatic halides, substituted and unsubstituted aromatic hydrocarbons, aliphatic ethers or combinations thereof. In certain embodiments, the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof. In some embodiments, the solvent is a mixture of acetonitrile and DMF. In certain embodiments, the solvent is a 9:1 mixture of acetonitrile:DMF.
  • Suitable bases for step S-5 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof. In certain embodiments, the base is diisopropylethylamine.
  • Suitable temperatures at which the reaction described in step S-5 may occur include about −20° C. to about 60° C. In certain embodiments, the temperature is about −10° C. to about 25° C. In some embodiments, the temperature is about −10° C.
  • While not wishing to be bound by any particular theory, it is believed that the order of reagent addition may be useful in reducing the formation of byproducts in step S-5. In certain embodiments a compound of formula F is added slowly to a compound of formula C.
  • It will be appreciated that certain reaction conditions may result in the formation of a regioisomer in step S-5 (i.e., reaction of endocyclic pyrazole nitrogen with a compound of formula F). In certain embodiments, the product of step S-5 is treated to remove the undesired regioisomer. In some embodiments, the product of step S-5 is re-slurried with a particular solvent or solvents to remove the regioisomer. In some embodiments, the product of step S-5 is re-slurried with t-butyl methyl ether to remove the regioisomer.
  • At step S-6, a compound of formula B is reacted in a suitable solvent with N-acetylhomopiperazine, optionally in the presence of a suitable base and/or catalyst, to produce compound A, 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide. One of ordinary skill in the art will recognize that when a compound of formula B is 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom. Examples of such iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof. While not wishing to be bound by any particular theory, it is believed that 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be a more reactive species in step S-6 than 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, due to the greater leaving group character of iodide over bromide.
  • Suitable solvents for step S-6 include aprotic solvents, aliphatic halides, aliphatic ethers or combinations thereof. In certain embodiments, the solvent is selected from dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof. In some embodiments, the solvent is acetone.
  • Suitable bases for step S-6 include tertiary amines such as pyridine, N-methylmorpholine, 1,4-diazabicyclo[2.2.2]octane, triethylamine (TEA), 1,8-diazabicyclo[5.4.0]undec-7-ene, diisopropylethylamine, and tetramethylethylenediamine; potassium carbonate, sodium bicarbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, tetrabutylammonium hydroxide, benzyltrimethylammonium hydroxide, triethylbenzylammonium hydroxide, 1,1,3,3-tetramethylguanidine, and combinations thereof. In certain embodiments, the base is potassium carbonate. In certain embodiments, the potassium carbonate is milled. In certain embodiments, the base is diisopropylethylamine.
  • In certain embodiments, the iodide source used in step S-6 is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the potassium iodide is milled. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B.
  • Suitable temperatures at which the reaction described in step S-6 may occur include about −20° C. to about 60° C. In certain embodiments, the temperature is about −10° C. to about 25° C. In some embodiments, the temperature is about 25° C.
  • In certain embodiments, as described in Example 5, an extractive workup step may be performed following step S-6. In certain embodiments, the organic phase comprises one or more organic solvents. In some embodiments, the organic solvents are ethanol, methyl tetrahydrofuran, dichloromethane, or a combination thereof. In certain embodiments, the solvents are ethanol and methyl tetrahydrofuran. In certain embodiments, the solvents are ethanol and dichloromethane. In some embodiments, the organic solvents are 5% ethanol in methyl tetrahydrofuran. In some embodiments, the organic solvents are 5% ethanol in dichloromethane.
  • In certain embodiments, dimer compounds may be formed as side products at step S-6. In such cases, it has been found that performing additional washes following HCl salt formation may be useful for reducing the presence of such dimer products.
  • At step S-7, compound A is reacted with hydrogen chloride, or an equivalent thereof, to form compound 1,5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrogen chloride.
  • Suitable solvents for step S-7 include polar solvents such as C1 to C4 alcohols (e.g. ethanol, methanol, 2-propanol), water, acetone or combinations thereof. In certain embodiments, the solvent is selected from ethanol, water, acetone, or combination thereof. In some embodiments, the solvent a mixture of acetone, water, and ethanol.
  • It will be appreciated that various types of absolute or denatured ethanol may be used in accordance with the present invention. In some embodiments, the ethanol is ethanol 1 L (denatured with 9% acetone). In some embodiments, the ethanol is ethanol 2B (denatured with 0.5% toluene).
  • One of ordinary skill in the art will appreciate that a number of suitable forms of hydrogen chloride can be used in step S-7 to produce the desired hydrochloride salt. Examples of such suitable forms include hydrogen chloride gas, aqueous solutions of hydrogen chloride, and solutions of hydrogen chloride in aliphatic ethers. In certain embodiments, the HCl is provided as an aqueous solution in acetone.
  • In certain embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.5-1.1 equivalents. In certain embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.8-1.0 equivalents. In some embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.93 equivalents.
  • It will be appreciated that the addition of excess HCl in step S-7 may result in the formation of a di-HCl salt. In certain embodiments, formation of such di-HCl salts is minimized or avoided by the addition of HCl in equivalents as described herein. In certain embodiments, formation of such di-HCl salts is minimized or avoided by careful, controlled addition of HCl.
  • Suitable temperatures at which the reaction described in step S-7 may occur include about −20° C. to about 60° C. In certain embodiments, the temperature is about −10° C. to about 35° C. In some embodiments, the temperature is about 25° C. to about 30° C.
  • As exemplified in Examples 3, 5 and 8, and without wishing to be bound by any particular theory, it is believed that the ternary solvent system of acetone, water, and ethanol, along with the number of equivalents of HCl used relative to compound of formula A, may be useful for obtaining the desired polymorph form I of compound I.
  • In certain embodiments, each of the aforementioned synthetic steps may be performed sequentially with isolation of each intermediate D, C, B, and A performed after each step. Alternatively, each of steps S-3, S-4, S-5, S-6 and S-7, as depicted in Scheme 2 above, may be performed in a manner whereby no isolation of one or more intermediates D, C, B, and A is performed. In certain embodiments, steps S-5, S-6, and S-7 are performed in sequence without any isolation of intermediates. In certain embodiments, steps S-6 and S-7 are performed in sequence without any isolation of intermediates. While not wishing to be bound by any particular theory, it is believed that such techniques may be useful in obtaining compounds of formulae B, A, and I in greater yield and purity.
  • In certain embodiments, compound A can be prepared according to Scheme 3 set forth below:
  • Figure US20090264648A1-20091022-C00012
  • wherein K is as defined above.
  • At step S-3a, an ester of formula G is coupled with N-acetylhomopiperazine to form an ester of formula H. In certain embodiments, R is optionally substituted group selected from C1-6 aliphatic. In some embodiments, R is C1-3 aliphatic. In some embodiments, R is methyl.
  • A suitable base may be used to facilitate the reaction between a compound of formula G and N-acetylhomopiperazine. Examples of such bases include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, potassium carbonate, and combinations thereof. In certain embodiments, the base is potassium carbonate.
  • One of ordinary skill in the art will recognize that when the LG2 group of a compound of formula G is bromine, the iodo analog may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom. Examples of such iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof.
  • In certain embodiments, the iodide source is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula G. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula G.
  • In certain embodiments, step S-3a is carried out in the presence of a suitable solvent. In certain embodiments, the solvent is selected from a polar aprotic solvent. Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetamide, acetone, or combination thereof. In some embodiments, the solvent is acetone.
  • At step S-4a, an ester of formula H is saponified with a suitable acid or base to provide carboxylic acid J. One of ordinary skill in the art will be aware of appropriate acids and bases that may be used. Such suitable bases include strong inorganic bases i.e., those that completely dissociate in water under formation of hydroxide anion. Examples of such bases include alkaline metals, alkaline earth metal hydroxides, and combinations thereof. In some embodiments, a suitable acid is a Lewis acid.
  • At step S-5a, carboxylic acid J is chlorinated to form acyl chloride K. In certain embodiments, the chlorination is facilitated by POCl3. In certain embodiments, step S-5a is carried out in the presence of a suitable solvent. In certain embodiments, the solvent is selected from a polar aprotic solvent. Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetamide, acetone, or combination thereof. In some embodiments, the solvent is dimethylacetamide.
  • At step S-6a, acyl chloride K is reacted with aryl aminopyrazole C to provide compound A. Compound A may then be used as described above and herein.
  • In certain embodiments, the present invention provides a method for preparing compound I-1′:
  • Figure US20090264648A1-20091022-C00013
  • wherein each of Ar, Y, Y′, T, and Ring A is as defined above and herein;
    comprising the steps of:
    (a) providing compound I-1:
  • Figure US20090264648A1-20091022-C00014
  • and
    (b) treating said compound I-1 with hydrochloric acid to form compound I-1′.
  • One of ordinary skill in the art will appreciate that a number of suitable forms of hydrogen chloride can be used to produce the desired hydrochloride salt. Examples of such suitable forms include hydrogen chloride gas, aqueous solutions of hydrogen chloride, and solutions of hydrogen chloride in aliphatic ethers. In certain embodiments, the HCl is provided as an aqueous solution in acetone. In certain embodiments, compound A is treated with hydrochloric acid in a solvent mixture of acetone, water, and ethanol.
  • In certain embodiments, the number of equivalents of HCl used relative to compound of formula I-1 is about 0.5-1.1 equivalents. In certain embodiments, the number of equivalents of HCl used relative to compound of formula I-1 is about 0.8-1.0 equivalents. In some embodiments, the number of equivalents of HCl used relative to compound of formula I-1 is about 0.93 equivalents.
  • In certain embodiments, the present invention provides a method for preparing compound I-1:
  • Figure US20090264648A1-20091022-C00015
  • wherein each of Ar, Y, Y′, T, and Ring A is as defined above and herein,
    comprising the steps of:
    (a) providing compound B′:
  • Figure US20090264648A1-20091022-C00016
  • wherein, LG2 is a suitable leaving group, and
    (b) treating said compound of formula B′ with a compound of formula G′:
  • Figure US20090264648A1-20091022-C00017
  • optionally in the presence of a suitable base and/or additive, to form compound I-1.
  • The LG2 group of formula B′ is a suitable leaving group, as defined and described herein. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. Examples of the LG2 group of formula B′ include Br, I, —OMs, —OTs, and —OTf.
  • One of ordinary skill in the art will recognize that when a compound of formula B′ is 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom. Examples of such iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof.
  • In certain embodiments, the iodide source is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B′. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B′.
  • A suitable base may be used to facilitate the reaction between a compound of formula B′ and a compound of formula G′. Examples of such bases include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, potassium carbonate, and combinations thereof. In certain embodiments, the base is diisopropylethylamine. In certain embodiments, the base is potassium carbonate.
  • In certain embodiments, the transformation of a compound of formula B′ to compound I-1 is performed in the presence of a suitable solvent. In certain embodiments, the solvent is selected from a polar aprotic solvent. Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof. In some embodiments, the solvent is acetone.
  • According to another embodiment, the present invention provides a method for preparing a compound of formula B′:
  • Figure US20090264648A1-20091022-C00018
  • wherein each of Ar, Y, Y′, T, LG2 and Ring A is as defined above and herein comprising the steps of:
    (a) providing compound C:
  • Figure US20090264648A1-20091022-C00019
  • and
    (b) treating said compound C′ in the presence of a suitable base with a compound of formula F′:
  • Figure US20090264648A1-20091022-C00020
  • wherein LG3 is a suitable leaving group, to form a compound of formula B′.
  • The LG2 group of formulae B′ and F′ is a suitable leaving group, as defined and described herein. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. In certain embodiments, the LG2 group of formulae B′ and F′ include Br, I, —OMs, —OTs, and —OTf.
  • The LG3 group of formula F′ is a suitable leaving group, and defined and described herein. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OR,
  • Figure US20090264648A1-20091022-C00021
  • wherein each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. Examples of LG3 groups of formula F′ include —OH, —OMe, —OEt, —Cl, —Br,
  • Figure US20090264648A1-20091022-C00022
  • and the like. In some embodiments, LG3 is —Cl.
  • In certain embodiments, the compound of formula F′ is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F′ is 5-bromovaleryl chloride.
  • Suitable bases to facilitate the transformation of compound C′ to a compound of formula B′ include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, and combinations thereof. In certain embodiments, the base is diisopropylethylamine.
  • In certain embodiments, the transformation of compound C′ to a compound of formula B′ is carried out in the presence of a suitable solvent. In certain embodiments, the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof. In some embodiments, the solvent is a mixture of acetonitrile and DMF. In certain embodiments, the solvent is a 9:1 mixture of acetonitrile:DMF.
  • In certain embodiments, the present invention provides a method for preparing compound I:
  • Figure US20090264648A1-20091022-C00023
  • comprising the steps of:
    (a) providing compound A:
  • Figure US20090264648A1-20091022-C00024
  • and
    (b) treating said compound A with hydrochloric acid to form compound I.
  • One of ordinary skill in the art will appreciate that a number of suitable forms of hydrogen chloride can be used to produce the desired hydrochloride salt. Examples of such suitable forms include hydrogen chloride gas, aqueous solutions of hydrogen chloride, and solutions of hydrogen chloride in aliphatic ethers. In certain embodiments, the HCl is provided as an aqueous solution in acetone. In certain embodiments, compound A is treated with hydrochloric acid in a solvent mixture of acetone, water, and ethanol.
  • In certain embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.5-1.1 equivalents. In certain embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.8-1.0 equivalents. In some embodiments, the number of equivalents of HCl used relative to compound of formula A is about 0.93 equivalents.
  • In certain embodiments, the present invention provides a method for preparing compound A:
  • Figure US20090264648A1-20091022-C00025
  • comprising the steps of:
    (a) providing compound B:
  • Figure US20090264648A1-20091022-C00026
  • wherein, LG2 is a suitable leaving group, and
    (b) treating said compound of formula B with N-acetylhompiperazine, optionally in the presence of a suitable base and/or additive, to form compound A.
  • The LG2 group of formula B is a suitable leaving group. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. Examples of the LG2 group of formula B include —Br, —I, —OMs, —OTs, and —OTf.
  • One of ordinary skill in the art will recognize that when a compound of formula B is 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide, 5-iodo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide may be generated in situ by the addition of an iodide source, affording exchange of an iodine atom with the bromine atom. Examples of such iodide sources include, but are not limited to, sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodides, or mixtures thereof.
  • In certain embodiments, the iodide source is sodium iodide. In certain embodiments, the iodide source is potassium iodide. In some embodiments, the iodide source is used in catalytic amounts ranging from about 0.1 to about 0.5 equivalents relative to a compound of formula B. In other embodiments, the iodide source is used stoichiometrically relative to a compound of formula B.
  • A suitable base may be used to facilitate the reaction between a compound of formula B and N-acetylhompiperazine. Examples of such bases include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, potassium carbonate, and combinations thereof. In certain embodiments, the base is diisopropylethylamine. In certain embodiments, the base is potassium carbonate.
  • In certain embodiments, the transformation of a compound of formula B to compound A is performed in the presence of a suitable solvent. In certain embodiments, the solvent is selected from a polar aprotic solvent. Exemplary solvents include dichloromethane, diethyl ether, acetonitrile, THF, NMP, N-methyl morpholine, dimethylacetaminde, acetone, or combination thereof. In some embodiments, the solvent is acetone.
  • According to another embodiment, the present invention provides a method for preparing a compound of formula B:
  • Figure US20090264648A1-20091022-C00027
  • comprising the steps of:
    (a) providing compound C:
  • Figure US20090264648A1-20091022-C00028
  • and
    (b) treating said compound C in the presence of a suitable base with a compound of formula F:
  • Figure US20090264648A1-20091022-C00029
  • to form a compound of formula B.
  • The LG2 group of formulae B and F is a suitable leaving group. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OMs, —OTs, and —OTf. Examples of the LG2 group of formulae B and F include —Br, —I, —OMs, —OTs, and —OTf.
  • The LG3 group of formula F is a suitable leaving group. Suitable leaving groups are well known in the art and include, but are not limited to, halogen, —OR,
  • Figure US20090264648A1-20091022-C00030
  • wherein each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. Examples of LG3 groups of formula F include —OH, —OMe, —OEt, —Cl, —Br,
  • Figure US20090264648A1-20091022-C00031
  • and the like. In some embodiments, LG3 is —Cl.
  • In certain embodiments, the compound of formula F is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride. In some embodiments, the compound of formula F is 5-bromovaleryl chloride.
  • Suitable bases to facilitate the transformation of compound C to a compound of formula B include pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, and combinations thereof. In certain embodiments, the base is diisopropylethylamine.
  • In certain embodiments, the transformation of compound C to a compound of formula B is carried out in the presence of a suitable solvent. In certain embodiments, the solvent is selected from diethyl ether, t-butyl methyl ether, THF, ethyl acetate, DMSO, DMF, NMP, acetonitrile, dichloromethane, benzene, toluene, or combination thereof. In some embodiments, the solvent is a mixture of acetonitrile and DMF. In certain embodiments, the solvent is a 9:1 mixture of acetonitrile:DMF.
  • In certain embodiments, the present invention provides a method for preparing compound C:
  • Figure US20090264648A1-20091022-C00032
  • comprising the steps of:
    (a) providing compound D:
  • Figure US20090264648A1-20091022-C00033
  • and
    (b) treating said compound of formula D with hydrazine, or an equivalent thereof, to form compound C.
  • In certain embodiments, the hydrazine equivalent used is a hydrate. In some embodiments, the hydrazine equivalent is a monohydrate.
  • In certain embodiments, the transformation of compound D to compound C is carried out in the presence of a suitable solvent. In certain embodiments, the solvent is selected from ethanol, toluene, dichloromethane, diethyl ether, THF, benzene or combination thereof. In some embodiments, the solvent is ethanol.
  • In another embodiment, the present invention provides a method for preparing compound D:
  • Figure US20090264648A1-20091022-C00034
  • comprising the steps of:
    (a) combining a compound of formula E:
  • Figure US20090264648A1-20091022-C00035
  • wherein, LG1 is a leaving group, with acetonitrile to form a mixture thereof, and;
    (b) treating said mixture with a suitable base to give compound D.
  • In certain embodiments, the base is selected the group consisting of NaH, LDA, NaHMDS, LHMDS, KHMDS, potassium t-amylate, BuLi, and NaOtBu. In some embodiments, the base is LHMDS.
  • The acetonitrile used in step (a) may be used in a range of equivalents relative to aromatic ester E. In certain embodiments, the equivalents of acetonitrile range from 0.5 to 50 equivalents. In certain embodiments, the equivalents of acetonitrile range from 2 to 10 equivalents. In some embodiments, the equivalents of acetonitrile range from 4 to 6 equivalents.
  • In certain embodiments, the transformation of a compound of formula E to compound D may be carried out in the presence of a suitable solvent. In some embodiments, the solvent is selected from toluene, acetonitrile, diethyl ether, t-butyl methyl ether, THF, benzene, dichloromethane or combinations thereof. In certain embodiments, no solvent is used.
  • According to another embodiment, the present invention provides compound I substantially free of N-acetylhompiperazine, 5-iodovaleryl chloride, 5-bromovaleryl chloride, 5-chlorovaleryl chloride, and compounds F-1, F-2, and F-3:
  • Figure US20090264648A1-20091022-C00036
  • Compounds F-1, F-2, and F-3 are impurities that may arise from steps S-1, S-2, S-5, or S-6 as described above and herein. “Substantially free,” as used herein, means that at least about 80% by weight of the desired compound is present. In other embodiments, at least about 92% by weight of a desired compound is present. In still other embodiments of the invention, at least about 99% by weight of a desired compound is present. Such impurities may be isolated from product mixtures by any method known to those skilled in the art, including liquid chromatography (LC).
  • In some embodiments, the present invention provides compound I having total impurities of less than 0.5%, less than 0.4%, or less than 0.3% by weight.
  • The present invention provides methods that provide compound I in substantially higher yields than described previously (U.S. Patent Application Ser. No. 60/880,629, filed Jan. 16, 2007).
  • As will be readily apparent to one skilled in the art, the unsubstituted ring nitrogen pyrazoles, as in the compounds of the present invention, are known to rapidly equilibrate in solution, as mixtures of both tautomers:
  • Figure US20090264648A1-20091022-C00037
  • for the compounds described above and herein, where only one tautomer is indicated, the other tautomer is also intended as within the scope of the present invention. Certain compound names may indicate one tautomer. For the compounds named above and herein, where only one tautomer is indicated by the compound name, the other tautomer is also intended as within the scope of the present invention.
  • Compounds of the invention can be in the form of free bases or acid addition salts, preferably salts with pharmaceutically acceptable acids.
  • Pharmacological activity of a representative group of compounds of formula I-1 was demonstrated in an in vitro assay utilizing cells stably transfected with the alpha 7 nicotinic acetylcholine receptor and cells expressing the alpha 1 and alpha 3 nicotinic acetylcholine receptors and 5HT3 receptor as controls for selectivity.
  • Compounds of formula I-1 may be provided according to the present invention in any of a variety of useful forms, for example as pharmaceutically acceptable salts, as particular crystal forms, etc. In some embodiments, prodrugs of compounds of formula I-1 are provided. Various forms of prodrugs are known in the art, for example as discussed in Bundgaard (ed.), Design of Prodrugs, Elsevier (1985); Widder et al. (ed.), Methods in Enzymology, vol. 4, Academic Press (1985); Kgrogsgaard-Larsen et al. (ed.); “Design and Application of Prodrugs”, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991); Bundgaard et al., Journal of Drug Delivery Reviews, 8:1-38 (1992); Bundgaard et al., J. Pharmaceutical Sciences, 77:285 et seq. (1988); and Higuchi and Stella (eds.), Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975).
  • As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.
  • EXEMPLIFICATION Experimental Procedures Synthesis of Compounds General
  • Unless otherwise specified all nuclear magnetic resonance spectra were recorded using a Varian Mercury Plus 400 MHz spectrometer equipped with a PFG ATB Broadband probe. HPLC-MS analyses were performed with a Waters 2795 separation module equipped with a Waters Micromass ZQ (ES ionisation) and Waters PDA 2996, using a Waters XTerra MS C18 3.5 μm 2.1×50 mm column.
  • Preparative HLPC was run using a Waters 2767 system with a binary Gradient Module Waters 2525 pump and coupled to a Waters Micromass ZQ (ES) or Waters 2487 DAD, using a Supelco Discovery HS C18 5.0 μm 10×21.2 mm column Gradients were run using 0.1% formic acid/water and 0.1% formic acid/acetonitrile with gradient 5/95 to 95/5 in the run time indicated in the Examples.
  • All column chromatography was performed following the method of Still, C.; J. Org Chem 43, 2923 (1978). All TLC analyses were performed on silica gel (Merck 60 F254) and spots revealed by UV visualisation at 254 nm and KMnO4 or ninhydrin stain.
  • When specified for array synthesis, heating was performed on a Buchi Syncore® system. All microwave reactions were performed in a CEM Discover oven.
  • Abbreviations Used Throughout the Experimental Procedures
    • AcOEt ethyl acetate
    • DCM dichloromethane
    • DCE 1,2-dichloroethane
    • DMEA N,N-dimethylethylamine
    • DMF N,N-dimethylformamide
    • DMSO, dmso dimethylsulphoxide
    • DMA N,N-dimethylacetamide
    • scx strong cation exchanger
    • TEA triethylamine
    • TFA trifluoroacetic acid
    • THF tetrahydrofuran
    • TLC thin layer chromatography
    • LC-MS liquid chromatography-mass spectrometry
    • HPLC high performance liquid chromatography
    General 3-amino-5-aryl/heteroaryl pyrazole synthesis
  • The 3-amino-5-aryl/heteroaryl pyrazoles used in the Examples were either commercially available or synthesised using the routes shown in the scheme below:
  • Figure US20090264648A1-20091022-C00038
  • General procedure for aryl/heteroaryl β-ketonitrile synthesis (A1):
  • Figure US20090264648A1-20091022-C00039
  • Aryl or heteroaryl methyl carboxylate were commercially available or were synthesized according to the following standard procedure: the aryl or heteroaryl carboxylic acid (32 mmol) was dissolved in MeOH (40 mL) and sulfuric acid (1 mL) was added. The mixture was refluxed overnight, after which the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated aqueous NaHCO3 solution. The organic phase was dried and evaporated under reduced pressure, and the crude was used without further purification.
  • To a solution of an aryl or heteroaryl methyl carboxylate (6.5 mmol) in dry toluene (6 mL) under N2, NaH (50-60% dispersion in mineral oil, 624 mg, 13 mmol) was carefully added. The mixture was heated at 80° C. and then dry CH3CN was added dropwise (1.6 mL, 30.8 mmol). The reaction was heated for 18 hours and generally the product precipitated from the reaction mixture as Na salt.
  • The reaction was then allowed to cool down to room temperature and the solid formed was filtered and then dissolved in water. The solution was then acidified with 2N HCl solution and at pH between 2-6 (depending on the ring substitution on the aryl/heteroaryl system) the product precipitated and was filtered off. If no precipitation occurred, the product was extracted with DCM.
  • After work-up, the products were generally used in the following step without further purification. The general yield was between 40 and 80%.
  • General Procedure for Aryl/Heteroaryl β-Ketonitrile Synthesis (Route A1bis):
  • Figure US20090264648A1-20091022-C00040
  • Aryl- or heteroaryl-carboxylic acid methyl esters are commercially available or were synthesized under the standard procedure, as described in general procedure A1.
  • To a solution of dry alkanenitrile in toluene (1 mmol/mL, 5 eq.) cooled down to −78° C. under nitrogen, a solution of n-butyllithium in n-hexane (1.6 N, 3.5 eq) was added dropwise. The mixture was left stirring at −78° C. for 20 minutes and then a solution of the aryl or heteroaryl methyl carboxylate in toluene (0.75 mmol/mL, 1 eq.) was added and the reaction allowed to reach room temperature. Upon reaction completion, after about 20 minutes, the mixture was cooled down to 0° C. and HCl 2 N was added to pH 2. The organic phase was recovered, dried over Na2SO4 and concentrated under reduced pressure, affording the title product which was generally used without further purification.
  • General Procedure for Aryl Aminopyrazole Synthesis (Route A2):
  • Figure US20090264648A1-20091022-C00041
  • To a solution of the β-ketonitrile (7.5 mmoL), in absolute EtOH (15 mL) hydrazine monohydrate (0.44 mL, 9.0 mmol) was added and the reaction was heated at reflux for 18 hrs. The reaction mixture was allowed to cool to room temperature and the solvent was evaporated under reduced pressure. The residue was dissolved in DCM and washed with water.
  • The organic phase was concentrated under reduced pressure to give a crude product that was purified by SiO2 column or by precipitation from Et2O.
  • Yields were generally between 65 and 90%.
  • Hydroxy-Aryl- or Hydroxy-Heteroaryl-Carboxylic Acid to Methyl Ester—General Procedure
  • 4-hydroxy-benzoic acid (usually 24.0 mmol) was dissolved in MeOH (50 mL) and sulfuric acid (1 mL/g substrate) was added. The mixture was refluxed overnight, after which the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated NaHCO3 to basic pH. The organic phase was dried and evaporated under reduced pressure, and the product was used without further purification. The yields were between 80 and 90%.
  • Hydroxy-Aryl- or Hydroxy-Heteroaryl-Carboxylic Acid Methyl Ester to F2Cho-Aryl- or Heteroarylcarboxylic Acid Methyl Ester-General Procedure
  • Under a N2 atmosphere, 4-hydroxy-benzoic acid methyl or ethyl ester (1.0 eq) and sodium chlorodifluoroacetate (1.2 eq) were dissolved in DMF (20-25 mL) in a two neck round bottom flask; potassium carbonate (1.2 eq) was added and the mixture was heated at 125° C. until complete conversion of the starting material was observed by LC-MS. The mixture was then diluted with water and extracted with DCM; the organic phase was dried and removed under reduced pressure, and the crude was purified through Si column to obtain the product (Yields from 20 to 70%).
  • The following Table 1 reports yields and analytical data obtained in the preparation of a series of F2CHO-aryl- or F2CHO-heteroaryl-carboxylic acid methyl esters prepared according to the general procedures described above
  • TABLE 1
    Starting
    material Methyl ester —OH Methyl ester —OCHF2
    3-Fluoro-4- C8H7FO3 C9H7F3O3
    hydroxy- Yield = 85% Yield = 66%
    benzoic acid 1H NMR (DMSO-d6) δ 1H NMR (DMSO-d6) δ 3.78 (3H,
    3.78 (3H, s), 7.00-7.05 (1H, m), s), 6.24 (1H, m), 7.61 (1H, m),
    7.60-7.65 (2H, m) 7.64 (1H, m), 10.89 (1H, bs)
    2,6-Difluoro-4- C8H6F2O3 C9H6F4O3
    hydroxy- Yield = 85% Yield = 34%
    benzoic acid 1H NMR (DMSO-d6) δ 1H NMR (DMSO-d6) δ 3.86 (3H,
    3.79 (s, 3H, s), 6.53 (2H, d, J = 10.8 Hz), s), 7.18-7.24 (2H, m), 7.42 (1H, t,
    11.13 (1H, s) J = 72.4 Hz).
    3,5-Dichloro-4- Commercially available C9H6Cl2F2O3
    hydroxy- Yield = 74%
    benzoic acid 1H NMR (DMSO-d6) δ 3.31 (3H,
    s), 7.22 (1H, t, J = 71.6 Hz),
    8.05 (2H, s).
    3-Chloro-4- Commercially available C9H7ClF2O3
    hydroxy- Yield = 85%
    benzoic acid 1H NMR (DMSO-d6) δ 3.85 (3H,
    s), 7.39 (1H, t, J = 72.4 Hz),
    7.50 (1H, t, J = 8.4 Hz), 7.82-7.89 (2H,
    m).
    4-Hydroxy-3- Commercially available C10H10F2O4
    methoxy- Yield = 85%
    benzoic acid 1H NMR (DMSO-d6) 3.84 (3H,
    s), 3.87 (3H, s); 7.22 (1H, t, J = 73.6 Hz),
    7.29 (1H, d, J = 8.4 Hz),
    7.57-7.60 (2H, m).
    4-Hydroxy-2- C9H10O3 C10H10F2O3
    methyl-benzoic Yield = 95% Yield = 85%
    acid 1H NMR (DMSO-d6) 1H NMR (DMSO-d6) 2.52 (3H, br
    2.43 (3H, br s), 3.72 (3H, s); s), 3.80 (3H, s); 7.07-7.13 (2H, m);
    6.61-6.64 (2H, m); 7.71-7.73 (1H, 7.34 (1H, t, J = 73.6 Hz), 7.89 (1H,
    m), 10.10 (1H, s). d, J = 8.8 Hz).
  • 3-Imidazo[1,2-a]pyridin-6-yl-3-oxo-propionitrile
  • The product was obtained starting from imidazo[1,2-a]pyridine-6-carboxylic acid methyl ester according to general procedure A1
  • Yield 39%
  • C10H7N3O Mass (calculated) [185]; (found) [M+H+]=186 [M−H]=184
  • LC Rt=0.23, 100% (3 min method)
  • 1H-NMR: (dmso-d6): 4.72 (2H, s), 7.61-7.65 (2H, m), 7.70 (1H, m), 8.07 (1H, s), 9.40 (s, 1H).
  • 5-Imidazo[1,2-a]pyridin-6-yl-1H-pyrazol-3-ylamine
  • The title compound was synthesized according to general procedure A2 starting from 3-imidazo[1,2-a]pyridin-6-yl-3-oxo-propionitrile
  • Yield: 84%
  • C10H9N5 Mass (calculated) [199]; (found) [M+1]=200
  • LCMS, (5 min method, RT=0.21 min,
  • NMR (1H, 400 MHz, MeOH-d4) 3.34 (s, 2H), 5.90 (br s, 1H), 7.57 (s, 1H), 7.63 (br s, 1H), 7.86 (s, 1H), 8.73 (s, 1H)
  • Chlorocynnamonitrile Synthesis (Route B1)
  • Figure US20090264648A1-20091022-C00042
  • POC3 (2 eq with respect to the aryl/heteroaryl acetophenone) were added dropwise to 4 molar equivalents of anhydrous DMF cooled down to 0° C., at such a rate that the temperature did not exceed 10° C. The acetophenone (1 eq) was then added dropwise and the reaction was allowed to reach room temperature.
  • The reaction was then stirred for further 30 min and then 0.4 mmol of hydroxylamine hydrochloride were added. The reaction was then heated up to 50° C., after which heating was removed and additional 4 eq. of hydroxylamine hydrochloride were added portionwise (at such a rate that the temperature never exceeded 120° C.). The reaction was then stirred until the temperature of the mixture spontaneously decreased to 25° C. Water (100 mL) were then added and the mixture was extracted with diethyl ether. The organic phase was dried over Na2SO4 and concentrated under reduced pressure. The crude product was used for the next step without further purification.
  • Aryl Aminopyrazole Synthesis (Route B2)
  • Figure US20090264648A1-20091022-C00043
  • To a solution of the chlorocynnamonitrile (0.5 mmol/mL, 1 eq) in absolute EtOH 2 eq of hydrazine monohydrate were added and the reaction was heated at reflux for 4 hrs. The reaction mixture was allowed to cool to room temperature and the solvent was evaporated under reduced pressure. The residue was triturated with Et2O, allowing to recover the title compound which was generally used without further purification.
  • 5-(2-Trifluoromethyl-phenyl)-2H-pyrazol-3-ylamine a) 3-Oxo-3-(2-trifluoromethyl-phenyl)-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 2-trifluoromethyl-benzoic acid methyl ester (3.1 g, 14.0 mmol, 1.0 eq). The crude was precipitated from HCl to give the title product as a yellow solid (2.8 g, yield: 94%).
  • C10H6F3NO
  • 1H-NMR (CD3OD): 4.90 (2H, br s); 7.52-7.86 (4H, m).
  • b) 5-(2-Trifluoromethyl-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude was purified through Si column (eluent: DCM) and dried to give the title product (0.6 g, 20% Yield).
  • C10H8F3N3
  • 5-(2,6-Dimethyl-phenyl)-2H-pyrazol-3-ylamine a) 3-(2,6-Dimethyl-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1), refluxing the mixture overnight and then for 2 h at 110° C. The crude product was extracted with DCM and used in the following step without further purification (2.2 g, yield: 76%).
  • C11H11NO
  • b) 5-(2,6-Dimethyl-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude was purified through Si column (eluent: DCM) and washed with water, extracted and dried to give the title product (0.25 g, yield 10%).
  • C11H13N3
  • 1H-NMR (CD3OD): 2.09-2.23 (6H, m); 7.04-7.12 (2H, m); 7.18-7.26 (2H, m).
  • 5-(2-Chloro-4-fluoro-phenyl)-2H-pyrazol-3-ylamine a) 3-(2-Chloro-4-fluoro-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 2-chloro-4-fluoro-benzoic acid methyl ester (0.7 g, 3.7 mmol, 1.0 eq). The crude product was extracted with DCM and used in the following step without further purification (0.4 g, yield: 60%).
  • C9H5ClFNO
  • b) 5-(2-Chloro-4-fluoro-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude was dissolved in DCM, washed with sat NaHCO3, extracted and dried to give the title product (0.12 g, yield 26%).
  • C9H7ClFN3
  • 1H-NMR (dmso-d6): 7.03-7.53 (4H, m).
  • 5-(5-tert-Butyl-thiophen-2-yl)-2H-pyrazol-3-ylamine a) 3-(5-tert-Butyl-thiophen-2-yl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 5-tert-Butyl-thiophene-2-carboxylic acid methyl ester (3.0 g, 15.0 mmol, 1.0 eq). The crude product was extracted with DCM and used in the following step without further purification (2.7 g, yield: 86%).
  • C11H13NOS
  • b) 5-(5-tert-Butyl-thiophen-2-yl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude was washed with water and precipitated to give the title product (2.7 g, yield 91%).
  • C11H15N3S
  • Mass (calculated) [221]; (found) [M+H+]=222.
  • LC Rt=2.53 min, 94% (10 min method)
  • 1H-NMR (dmso-d6): 1.26-1.29 (9H, m); 4.87 (2H, br s); 5.47 (1H, br s); 6.66-6.79 (1H, m); 6.97-7.02 (1H, m)
  • 5-(3-Chloro-2-methyl-phenyl)-2H-pyrazol-3-ylamine a) 2-Ethyl-benzoic acid methyl ester
  • 2-Ethyl-benzoic acid (3.0 g, 17.6 mmol) was dissolved in MeOH (20 mL) and sulfuric acid (1 mL) was added. The mixture was refluxed overnight, after which the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated Na2CO3 to basic pH. The organic phase was dried and evaporated under reduced pressure, and the product (3.1 g, yield 96%) was used without further purification
  • C9H9ClO2
  • 1H-NMR (dmso-d6): 2.48 (3H, br s); 3.82 (3H, s); 7.31 (1H, t, J=7.6 Hz); 7.63-7.67 (2H, m).
  • b) 3-(3-Chloro-2-methyl-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 3-Chloro-2-methyl-benzoic acid methyl ester (3.1 g, 16.8 mmol, 1.0 eq). The crude product was precipitated form water and used in the following step without further purification (2.4 g, yield: 74%).
  • C10H8ClNO
  • 1H-NMR (dmso-d6): 2.31 (3H, br s); 4.64 (2H, br s); 7.27-7.36 (2H, m); 7.54-7.77 (1H, m).
  • c) 5-(3-Chloro-2-methyl-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column (20 g) with gradient elution from 100% EtOAc to EtOAc-MeOH 80:20. The title product (1.3 g, yield 50%) was obtained.
  • C10H10ClN3
  • Mass (calculated) [207]; (found) [M+H+]=208.
  • LC Rt=1.96 min, 85% (10 min method)
  • 1H-NMR (CDCl3): 2.41 (3H, s); 5.74 (1H, s); 7.16 (1H, t, J=8.0 Hz); 7.20-7.26 (1H, m); 7.38-7.40 (1H, m).
  • 5-(2-Ethyl-phenyl)-2H-pyrazol-3-yl-amine a) 2-Ethyl-benzoic acid methyl ester
  • 2-Ethyl-benzoic acid (3.0 g, 20.0 mmol) was dissolved in MeOH (20 mL) and catalytic quantity of sulfuric acid (1 mL) was added. The mixture was refluxed overnight, after that the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated Na2CO3 to basic pH. The organic phase was dried and evaporated under reduced pressure, and the product (2.9 g, yield 88%) was used without further purification
  • C10H12O2
  • 1H-NMR (dmso-d6): 1.12 (3H, t, J=7.2 Hz); 2.86 (2H, q, J=7.2 Hz); 3.81 (3H, s); 7.27-7.34 (2H, m); 7.46-7.51 (1H, m); 7.73-7.75 (1H, m).
  • b) 3-(2-Ethyl-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 2-ethyl-benzoic acid methyl ester (2.9 g, 17.6 mmol, 1.0 eq). The crude product was extracted with DCM as a yellow oil and used in the following step without further purification (2.8 g, yield: 92%).
  • C11H11NO
  • 1H-NMR (dmso-d6): 1.10-1.18 (3H, m); 2.78 (2H, q, J=7.2 Hz); 4.67 (1H, s); 7.23-7.53 (3H, m); 7.73-7.78 (1H, m).
  • c) 5-(2-Ethyl-phenyl)-2H-pyrazol-3-yl-amine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column (20 g) with gradient elution from 100% EtOAc to EtOAc-MeOH 80:20. The title product (1.2 g, yield 40%) was obtained
  • C11H13N3
  • Mass (calculated) [187]; (found) [M+H+]=188.
  • LC Rt=1.58 min, 90% (10 min method)
  • 1H-NMR (CDCl3): 1.15 (3H, t, J=7.6 Hz); 2.71 (2H, q, J=7.6 Hz); 5.72 (1H, s); 7.20-7.26 (1H, m); 7.29-7.35 (3H, m).
  • 5-(4-Methoxy-phenyl)-4-methyl-2H-pyrazol-3-ylamine a) 3-(4-Methoxy-phenyl)-2-methyl-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 4-methoxy-benzoic acid methyl ester (3.0 mL, 18.0 mmol, 1.0 eq), NaH (1.4 g, 36.0 mmol, 2.0 eq) and propionitrile (6.1 mL, 84.9 mmol, 4.7 eq). The crude was purified through Si-column (eluent hexane/ethyl acetate) to give 2.1 g of title product (yield: 62%).
  • C11H11NO2
  • b) 5-(4-Methoxy-phenyl)-4-methyl-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was washed with basic water and dried, and the title product (1.8 g, yield 80%) was used without further purification
  • C11H13N3O
  • Mass (calculated) [203]; (found) [M+H+]=204.
  • LC Rt=1.34 min, 91% (10 min method)
  • 1H-NMR (CDCl3): 2.03 (3H, s); 3.84 (3H, s); 6.96-6.98 (2H, m); 7.37-7.39 (2H, m).
  • 4-Methyl-5-(4-trifluoromethyl-phenyl)-2H-pyrazol-3-ylamine a) 2-Methyl-3-oxo-3-(4-trifluoromethyl-phenyl)-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from 4-trifluoromethyl-benzoic acid methyl ester (3.0 g, 14.7 mmol, 1.0 eq), NaH (1.2 g, 29.4 mmol, 2.0 eq) and propionitrile (4.9 mL, 69.4 mmol, 4.7 eq). The crude product was extracted with DCM and used in the following step without further purification (3.2 g, yield: 96%).
  • C11H8F3NO
  • b) 4-Methyl-5-(4-trifluoromethyl-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was washed with basic water and dried, and the title product (2.8 g, yield 84%) was used without further purification
  • C11H10F3N3
  • Mass (calculated) [241]; (found) [M+H+]=242.
  • LC Rt=2.34 min, 92% (10 min method)
  • 1H-NMR (CDCl3): 2.05 (3H, s); 7.56 (2H, d, J=8.4 Hz); 7.64 (2H, d, J=8.4 Hz).
  • 5-(4-Cyclopropylmethoxy-2-methyl-phenyl)-2H-pyrazol-3-ylamine a) 4-Hydroxy-2-methyl-benzoic acid methyl ester
  • 4-Hydroxy-2-methyl-benzoic acid (4.8 g, 32.0 mmol) was dissolved in MeOH (40 mL) and catalytic quantity of sulfuric acid (1 mL) was added. The mixture was refluxed overnight, after which the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated NaHCO3 to basic pH. The organic phase was dried and evaporated under reduced pressure, and the product (5.0 g, yield 95%) was used without further purification.
  • C9H10O3
  • 1H-NMR (dmso-d6): 2.43 (3H, s); 3.72 (3H, s); 6.62-6.64 (2H, m); 7.71-7.73 (1H, m); 10.10 (1H, s).
  • b) 4-Cyclopropylmethoxy-2-methyl-benzoic acid methyl ester
  • 4-Hydroxy-2-methyl-benzoic acid methyl ester (1.0 g, 6.0 mmol, 1.0 eq) was dissolved in acetone (14 mL), NaI (0.45 g, 3.0 mmol, 0.5 eq) and K2CO3 (1.66 g, 12.0 mmol, 2.0 eq) were added ad the mixture was stirred at room temperature for 20 min. (Bromomethyl)cyclopropane (0.53 mL, 5.4 mmol, 0.9 eq) was added, and the mixture was refluxed for 2 days. The solvent was concentrated under reduced pressure, NaOH 10% was added, and the crude was extracted with DCM and dried. 0.42 g of title product (yield 32%) were recovered and used without further purification.
  • C13H16O3
  • 1H-NMR (CDCl3): 0.23-0.34 (2H, m); 0.52-0.64 (2H, m); 1.15-1.24 (1H, m); 2.52 (3H, s); 3.75 (2H, d, J=7.2 Hz); 3.77 (3H, s); 6.64-6.66 (1H, m); 7.83-7.85 (2H, m).
  • c) 3-(4-Cyclopropylmethoxy-2-methyl-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis from 4-cyclopropylmethoxy-2-methyl-benzoic acid methyl ester (route A1bis). 0.54 g of the title product was extracted from water and dried (yield 69%) and used directly for the next step.
  • C14H15NO2
  • d) 5-(4-Cyclopropylmethoxy-2-methyl-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column with gradient elution from 100% EtOAc to EtOAc-MeOH 90:10. The title product (206 mg, yield 36%) was obtained.
  • C14H17N3O
  • 1H-NMR (CD3OD): 0.29-0.36 (2H, m); 0.54-0.63 (2H, m); 1.18-1.28 (1H, m); 2.33 (3H, s); 3.81 (2H, d, J=7.2 Hz); 5.67 (1H, s); 6.74-6.80 (2H, m); 7.25 (1H, d, J=8.8 Hz).
  • 5-(3-Chloro-4-cyclopropylmethoxy-phenyl)-2H-pyrazol-3-ylamine a) 3-Chloro-4-cyclopropylmethoxy-benzoic acid methyl ester
  • 3-Chloro-4-hydroxy-benzoic acid methyl ester (1.1 g, 6.0 mmol, 1.0 eq) was dissolved in acetone (14 mL), NaI (0.45 g, 3.0 mmol, 0.5 eq) and K2CO3 (1.66 g, 12.0 mmol, 2.0 eq) were added ad the mixture was stirred at room temperature for 20 min. (Bromomethyl)cyclopropane (0.53 mL, 5.4 mmol, 0.9 eq) was added, and the mixture was refluxed for 2 days. The solvent was concentrated under reduced pressure, NaOH 10% was added, and the crude was extracted with DCM and dried. The title product (0.88 g, yield 32%) was recovered and used without further purification.
  • C12H13C3
  • 1H-NMR (dmso-d6): 0.33-0.37 (2H, m); 0.55-0.60 (2H, m); 1.25-1.27 (1H, m); 3.80 (3H, s); 3.99 (2H, d, J=7.2 Hz); 7.21 (1H, s, J=8.8 Hz); 7.85-7.91 (2H, m).
  • b) 3-(3-Chloro-4-cyclopropylmethoxy-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure from 3-Chloro-4-cyclopropylmethoxy-benzoic acid methyl ester (route A1bis). 0.74 g of the title product was extracted from water and dry (yield 81%) and used directly for the next step.
  • C13H12ClNO2
  • c) 5-(3-Chloro-4-cyclopropylmethoxy-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column (gradient elution from 100% EtOAc to EtOAc-MeOH 90:10). 521 mg of the title product (yield 67%) were obtained.
  • C13H14ClN3O
  • Mass (calculated) [263]; (found) [M+H+]=264.
  • LC Rt=2.51 min, 90% (10 min method)
  • 1H-NMR (CD3OD): 0.25-0.29 (2H, m); 0.52-0.55 (2H, m); 1.10-1.18 (1H, m); 3.81 (2H, d, J=6.8 Hz); 5.74 (1H, s); 6.95-6.99 (1H, m); 7.24-7.30 (2H, m).
  • 5-(4-Cyclopropylmethoxy-2-trifluoromethyl-phenyl)-2H-pyrazol-3-ylamine a) 4-hydroxy-2-trifluoromethyl-benzoic acid methyl ester
  • 4-hydroxy-2-trifluoromethyl-benzoic acid (5.0 g, 24.0 mmol) was dissolved in MeOH (50 mL) and a catalytic quantity of sulfuric acid was added. The mixture was refluxed overnight, after which the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated NaHCO3. The organic phase was dried and evaporated under reduced pressure, and the product was used without further purification.
  • C9H7F3O3
  • b) 4-Cyclopropylmethoxy-2-trifluoromethyl-benzoic acid methyl ester
  • 4-hydroxy-2-trifluoromethyl-benzoic acid methyl ester (1.1 g, 4.8 mmol, 1.0 eq) was dissolved in acetone (14 mL), NaI (0.5 eq) and K2CO3 (1.04 g, 2.0 eq) were added and the mixture was stirred at room temperature for 30 min. (Bromomethyl)cyclopropane (0.42 mL, 4.3 mmol, 0.9 eq) was added, and the mixture was refluxed for 2 days. The solvent was concentrated under reduced pressure, NaOH 10% was added, and it was extracted with DCM and dried. The title product (1.21 g, yield 92%) was recovered and used without further purification.
  • C13H13F3O3
  • c) 3-(4-Cyclopropylmethoxy-2-trifluoromethyl-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl IM and the organic phase separated and dried, to give 1.2 g of the title product (yield 94%) which was used directly for the next step.
  • C14H12F3NO2
  • Mass (calculated) [283]; (found) [M+H+]=284
  • LC Rt=3.86 min, 98% (10 min method)
  • d) 5-(4-Cyclopropylmethoxy-2-trifluoromethyl-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column (gradient elution from Ethyl Acetate-cycloexane 1:1 to Ethyl Acetate-MeOH 90:10). 650 mg of the title product (yield 52%) were obtained.
  • C14H14F3N3O
  • Mass (calculated) [297]; (found) [M+H+]=298.
  • LC Rt=2.78 min, 59% (10 min method)
  • 1H-NMR (CDCl3): 032-0.44 (2H, m); 0.64-0.62 (2H, m); 1.22-1.37 (1H, m); 3.80-3.92 (2H, m); 5.78 (1H, s); 7.04-7.07 (1H, m); 7.24-7.26 (1H, m); 7.38-7.40 (1H, m)
  • 5-(4-Cyclopropylmethoxy-2,3-difluoro-phenyl)-2H-pyrazol-3-ylamine a) 4-hydroxy-2,3-difluoro-benzoic acid methyl ester
  • 4-hydroxy-2,3-difluoro-benzoic acid (2.0 g, 11.5 mmol) was dissolved in MeOH (20 mL) and catalytic quantity of sulfuric acid was added. The mixture was refluxed overnight, after that the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated NaHCO3. The organic phase was dried and evaporated under reduced pressure, and the product was used without further purification.
  • C8H6F2O3
  • b) 4-Cyclopropylmethoxy-2,3-difluoro-benzoic acid methyl ester
  • 4-Hydroxy-2,3-difluoro-benzoic acid methyl ester (0.9 g, 4.8 mmol, 1.0 eq) was dissolved in acetone (14 mL), NaI (0.5 eq) and K2CO3 (1.03 g, 2.0 eq) were added and the mixture was stirred at room temperature for 30 min. (Bromomethyl)cyclopropane (0.42 mL, 0.9 eq) was added, and the mixture was refluxed for 2 days. The solvent was concentrated under reduced pressure, NaOH 10% was added, and it was extracted with DCM and dried. The title product (0.97 g, yield 84%) was recovered and used without further purification.
  • C12H12F2O3
  • c) 3-(4-Cyclopropylmethoxy-2,3-difluoro-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl IM and the organic phase separated and dried, to give 0.79 g of the title product (yield 79%) which was used directly for the next step.
  • C13H11F2NO2
  • Mass (calculated) [251]; (found) [M+H+]=252.
  • LC Rt=3.53 min, 82% (10 min method)
  • d) 5-(4-Cyclopropylmethoxy-2,3-difluoro-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column (gradient elution from EtOAc-cycloexane 1:1 to EtOAc:MeOH 90:10). 810 mg of the title product (yield 97%) were obtained.
  • C13H13F2N3O
  • Mass (calculated) [265]; (found) [M+H+]=266.
  • LC Rt=2.59 min, 75% (10 min method)
  • 1H-NMR (CDCl3): 032-0.47 (2H, m); 0.64-0.75 (2H, m); 1.19-1.38 (1H, m); 3.67-4.15 (4H, m); 5.95 (1H, s); 6.74-6.88 (1H, m); 7.17-7.26 (1H, m);
  • 5-(3,5-Dichloro-4-cyclopropylmethoxy-phenyl)-2H-pyrazol-3ylamine a) 3,5-Dichloro-4-Cyclopropylmethoxy-benzoic acid methyl ester
  • 3,5-Dichloro-4-hydroxy-benzoic acid ethyl ester (1.0 g, 4.5 mmol, 1.0 eq) was dissolved in acetone (14 mL), NaI (0.5 eq) and K2CO3 (0.98 g, 9.0 mmol, 2.0 eq) were added ad the mixture was stirred at room temperature for 30 min. (Bromomethyl)cyclopropane (0.39 mL, 4.1 mmol, 0.9 eq) was added, and the mixture was refluxed for 2 days. The solvent was concentrated under reduced pressure, NaOH 10% was added, and it was extracted with DCM and dried. The title product (0.98 g, yield 79%) was recovered and used without further purification.
  • C12H12Cl2O3
  • b) 3(3,5-Dichloro-4-cyclopropylmethoxy-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl 1M and the organic phase separated and dried, to give 0.91 g of the title product (yield 90%) which was used directly for the next step.
  • C13H13Cl2N3O
  • Mass (calculated) [283]; (found) [M+H+]=284.
  • LC Rt=4.06 min, 99% (10 min method)
  • c) 5-(3,5-Dichloro-4-cyclopropylmethoxy-phenyl)-2H-pyrazol-3ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column (gradient elution from EtOAc-cycloexane 1:1 to Ethyl Acetate:MeOH 90:10). 750 mg of the title product (yield 79%) were obtained.
  • C13H13Cl2N3O
  • Mass (calculated) [297]; (found) [M+H+]=298.
  • LC Rt=3.23 min, 93% (10 min method)
  • 1H-NMR (CDCl3): 023-0.46 (2H, m); 0.64-0.74 (2H, m); 1.30-1.48 (1H, m); 3.60-4.04 (4H, m); 5.86 (1H, s); 7.48 (2H, s)
  • 5-(4-Cyclopropylmethoxy-3-methoxy-phenyl)-2H-pyrazol-3-ylamine a) 4-Cyclopropylmethoxy-3-methoxy-benzoic acid methyl ester
  • 4-hydroxy-3-methoxy-benzoic acid methyl ester (1.0 g, 5.5 mmol, 1.0 eq) was dissolved in acetone (14 mL), NaI (0.5 eq) and K2CO3 (1.0 g, 2.0 eq) were added and the mixture was stirred at room temperature for 30 min. (Bromomethyl)cyclopropane (0.53 mL, 0.9 eq) was added, and the mixture was refluxed for 2 days. The solvent was concentrated under reduced pressure, NaOH 10% was added, and it was extracted with DCM and dried. The title product (1.21 g, yield 93%) was recovered and used without further purification.
  • C13H16O4
  • b) 3(4-Cyclopropylmethoxy-3-methoxy-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure (route A1bis). The mixture was acidified with HCl IM and the organic phase separated and dried, to give 1.24 g of the title product (yield 99%) which was used directly for the next step.
  • C14H15NO3
  • Mass (calculated) [245]; (found) [M+H+]=246.
  • LC Rt=3.03 min, 100% (10 min method)
  • c) 5-(4-Cyclopropylmethoxy-3-methoxy-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through SiO2 column (gradient elution from EtOAc-cycloexane 1:1 to Ethyl Acetate:MeOH 90:10). 220 mg of the title product (yield 50%) were obtained.
  • C14H17N3O2
  • Mass (calculated) [259]; (found) [M+H+]=260.
  • LC Rt=1.86 min, 93% (10 min method)
  • 1H-NMR (CDCl3): 027-0.43 (2H, m); 0.56-0.72 (2H, m); 1.23-1.40 (1H, m); 348 (2H, m); 3.87 (3H, s); 3.98 (2H, br s); 5.82 (1H, s); 6.85-6.89 (1H, m); 7.05-7.10 (2H, m);
  • 3-Amino-5-(3-fluoro-phenyl)-pyrazole-1-carboxylic acid tert-butyl ester
  • 3-Amino-5-(3-fluoro-phenyl)-pyrazole (5.0 g, 28.0 mmol, 1.0 eq) and KOH 4.5 M (50 mL, 226 mmol, 8 eq) were dissolved in DCM (200 mL), and di-tert-butyl dicarbonate (6.5 g, 30.0 mmol, 1.1 eq) was added; the mixture was stirred at room temperature until complete conversion was observed by LC-MS analysis. The organic phase was washed with saturated brine and evaporated; the crude was crystallized with MeOH, to give 7.4 g of title product (yield 95%).
  • C14H16FN3O2
  • 1H-NMR (dmso-d6): 1.57 (9H, s), 5.80 (1H, s), 6.43 (2H, br s), 7.16-7.21 (1H, m), 7.41-7.47 (1H, m); 7.50-7.54 (1H, m); 7.58-7.60 (1H, m).
  • 3-Amino-5-o-tolyl-pyrazole-1-carboxylic acid tert-butyl ester
  • 3-Amino-5-o-tolyl-pyrazole (0.5 g, 2.89 mmol, 1.0 eq) and KOH 4.5 M (5.1 mL, 23.1 mmol, 8.0 eq) were dissolved in DCM (20 mL), and Di-tert-butyl dicarbonate (0.66 g, 3.0 mmol, 1.1 eq) was added; the mixture was stirred at room temperature until complete conversion was observed by LC-MS analysis. The organic phase was washed with saturated brine and evaporated, to give 0.6 g of title product (yield 76%).
  • C15H19N3O2
  • Mass (calculated) [273]; (found) [M+H+]=274.
  • LC Rt=2.34 min, 96% (5 min method)
  • 3-Amino-5-(4-trifluoromethyl-phenyl)-pyrazole-1-carboxylic acid tert-butyl ester
  • 3-Amino-5-(4-trifluoromethyl-phenyl)-pyrazole (2.0 g, 8.8 mmol, 1.0 eq) and KOH 4.5 M (15.7 mL, 70.5 mmol, 8.0 eq) were dissolved in DCM (70 mL), and di-tert-butyl dicarbonate (2.02 g, 9.2 mmol, 1.1 eq) was added; the mixture was stirred at room temperature until complete conversion was observed by LC-MS analysis. The organic phase was washed with saturated brine and evaporated; the crude was crystallized with CH3CN, to give 1.9 g of title product (yield 69%).
  • Mass (calculated) [327]; (found) [M+H+]=328.
  • LC Rt=2.59 min, 100% (5 min method)
  • 1H-NMR (dmso-d6): 1.57 (9H, s), 5.83 (1H, s), 6.46 (2H, s), 7.74 (2H, d, J=8.4 Hz), 7.95 (2H, d, J=8.8 Hz)
  • 5-Pyridin-2-yl-2H-pyrazol-3-ylamine a) Oxo-pyridin-2-yl-acetonitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1) from pyridine-2-carboxylic acid methyl ester (3.0 g, 21.9 mmol, 1.0 eq). The crude was precipitated from HCl to give the title product as a solid (2.2 g, yield: 69%) which was used directly for the next step.
  • C8H6N2O
  • b) 5-Pyridin-2-yl-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was dissolved in EtOAc, washed with NaHCO3, dried and evaporated. NMR analysis showed that a major portion of the crude mixture was still in the opened form: the mixture was then dissolved in CH3COOH and heated at 80° C. overnight, to allow for ring closure of the opened form. The product was then recovered as the acylated form, which was de-acylated stirring with HCl 6N at 60° C. overnight obtaining the title product (0.816 g, yield 60%).
  • C8H8N4
  • 1H-NMR (dmso-d6): 4.81 (2H, bs), 5.92 (1H, s), 7.21-7.24 (1H, m), 7.76 (2H, d), 8.51 (1H, d), 11.96 (1H, bs)
  • 5-(3-Difluoromethoxy-phenyl)-2H-pyrazol-3-ylamine a) 3-Difluoromethoxy-benzoic acid methyl ester
  • Difluoromethoxy-benzoic acid (2.0 g, 10.6 mmol, 1.0 eq) was dissolved in MeOH (15 mL) and a catalytic quantity of sulfuric acid was added. The mixture was refluxed overnight, after which the solvent was evaporated under reduced pressure; the crude was dissolved in DCM and washed with saturated NaHCO3 to basic pH. The organic phase was dried and evaporated under reduced pressure, and the title product was used without further purification (1.9 g, yield 90%).
  • C9H8F2O3
  • 1H-NMR (dmso-d6): 3.86 (3H, s), 7.33 (1H, t, J=73.6 Hz), 7.46-7.50 (1H, m), 7.59 (1H, t, J=8.0 Hz), 7.67 (1H, s); 7.82 (1H, d, J=7.6 Hz).
  • b) 3-(3-Difluoromethoxy-phenyl)-3-oxo-propionitrile
  • The product was prepared according to the general procedure for aminopyrazole synthesis (route A1 bis) from 3-difluoromethoxy-benzoic acid methyl ester (1.5 g, 7.4 mmol, 1.0 eq). The crude was precipitated by addition of aqueous HCl to give the product which was used directly for the next step.
  • C10H7F2NO2
  • c) 5-(3-Difluoromethoxy-phenyl)-2H-pyrazol-3-ylamine
  • The product was prepared according to general procedure for aminopyrazole synthesis (route A2). The crude product was purified through Si-column with gradient elution from 100% EtOAc to EtOAc-MeOH 90:10. 1.45 g of title product (yield 87%) was obtained.
  • C10H9F2N3O
  • 1H-NMR (dmso-d6): 4.89 (2H, br s), 5.75 (1H, s), 7.02 (1H, d), 7.25 (1H, t, J=74.0 Hz), 7.36-7.42 (2H, m), 7.48-7.50 (1H, d), 11.76 (1H, br s)
  • 5-Pyrazolo[1,5-a]pyridin-3-yl-2H-pyrazol-3-ylamine a) 3-Oxo-3-pyrazolo[1,5-a]pyridin-3-yl-propionitrile
  • To a solution of dry acetonitrile in toluene (0.66 mL, 13 mmol, 5 eq) cooled down to −78° C. under nitrogen, a solution of n-butyllithium in n-hexane (5.2 mL, 13 mmol, 5 eq) was added dropwise. The mixture was left stirring at −78° C. for 20 minutes and then a solution of pyrazolo[1,5-a]pyridine-3-carboxylic acid methyl ester (0.46 g, 2.6 mmol, 1 eq, prepared according to the reported procedure (Anderson et al. Journal of Heterocyclic Chemistry 1981, 18, 1149-1152) in toluene was added and the reaction allowed to reach room temperature. Upon reaction completion, after about 20 minutes, the mixture was cooled down to 0° C. and HCl 2N was added to pH 2. The organic phase was recovered, dried over Na2SO4 and concentrated under reduced pressure, affording the title product which was used without further purification in the following step.
  • C10H7N3O
  • b) 5-Pyrazolo[1,5-a]pyridin-3-yl-2H-pyrazol-3-ylamine
  • To a solution of the 3-oxo-3-pyrazolo[1,5-a]pyridin-3-yl-propionitrile (0.66 g, 3.6 mmol), in absolute EtOH (25 mL) hydrazine monohydrate (0.44 mL, 9.0 mmol) was added and the reaction was heated at reflux for 18 hours. The reaction mixture was allowed to cool to room temperature and the solvent was evaporated under reduced pressure. The residue was dissolved in DCM and washed with water.
  • The organic phase was concentrated under reduced pressure to give a crude product that was purified by SiO2 column (DCM to DCM:MeOH 95:5 to 85:15 gradient), yielding the title compound in 41% Yield (0.29 g, 1.48 mmol).
  • C10H9N5
  • 1H-NMR (dmso-d6): 8.68 (s, 1H); 8.21 (s, 1H); 7.92 (s, 1H); 7.28 (s, 1H); 6.90 (s, 1H); 5.75 (s, 1H); 5.10 (s, 2H).
  • Mass (calculated) [199]; (found) [M+H+]=200.
  • LC Rt=0.86 min, 92% (5 min method).
  • Example 1 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide i) 5-Bromo-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide
  • A solution of 5-bromovaleryl chloride (2.1 mL, 15.7 mmol, 1 eq) in dry DMA (35 mL) was cooled to −10° C. (ice/water bath) under N2; a solution of 5-(4-methoxy-phenyl)-1H-pyrazol-3-ylamine (3.0 g, 15.7 mmol, 1 equiv.) and diisopropylethylamine (2.74 mL, 15.7 mmol, 1 equiv.) in dry DMA (15 mL) was added over 30 min. After 2 hrs at −10° C., LC-MS shows completion of the reaction which was quenched by addition of H2O (ca. 50 mL). The solid which precipitates was filtered and washed with Et2O, to give 4.68 g of 5-bromo-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide as a white powder (13.3 mmol, 85% yield).
  • mp=149.5-151.5° C.
  • C15H18BrN3O2 Mass (calculated) [352.23]; (found) [M+H+]=352.09/354.10
  • LC Rt=2.07, 95% (5 min method)
  • 1H-NMR (400 MHz, DMSO-d6): δ 1.69-1.63 (2H, m); 1.81-1.75 (2H, m); 2.29 (2H, t); 3.52 (2H, t); 3.75 (3H, s); 6.75 (1H, bs); 6.96 (2H, d); 7.6 (2H, d); 10.28 (1H, s); 12.57 (1H, s)
  • ii) 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide
  • To 750 mg (1.96 mmol) of 5-bromo-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide in 7 mL of DMA, N-acetyl-diazepine (278 mg, 1.96 mmol) and NaI (240 mg, 1.96 mmol) were added and the reaction heated at 60° C. for 18 hours. Upon complete conversion (as monitored by LC-MS) the mixture was diluted with 20 mL of DCM and washed with water. The organic phase was concentrated under reduced pressure to afford a residue which was purified with a SiO2 column (10 g) eluting with a gradient from DCM to MeOH 90:10. The title compound (380 mg) was recovered pure (yield 46%).
  • C22H31N5O3 Mass (calculated) [413]; (found) [M+H+]=414
  • LC Rt=1.91, 100% (10 min method)
  • 1H-NMR (400 MHz, DMSO-d6): δ 1.53-1.75 (4H, m), 1.90-2.15 (5H, m), 2.28-2.42 (2H, m), 2.90-3.26 (3H, m), 3.34-3.58 (3H, m), 3.71-3.88 (7H, m)
  • Example 2 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide (mono hydrochloride salt)
  • To a solution of 5-(4-methoxyphenyl)-1H-pyrazol-3-ylamine (12 g, 62.8 mmol) and N,N-diisopropylethylamine (10.96 mL, 62.8 mmol) in dry N,N-dimethylformamide (150 mL) at −10° C. was added a solution of 5-bromovaleryl chloride (8.4 mL, 62.8 mmol) in dry N,N-dimethylformamide (50 mL) slowly (˜40 min) and the reaction mixture was allowed to stir at −10 to 0° C. for 8 hrs. Sodium iodide (9.44 g, 62.8 mmol) was added at 0° C. and followed by N-acetylhomopiperazine (8.24 mL, 62.8 mmol) and N,N-diisopropylethylamine (10.96 mL, 62.8 mmol) and the reaction mixture was allowed to stir at 50° C. for 18 hrs. The solvent was removed in vacuo. The residue was dissolved in methylene chloride (500 mL) and saturated aqueous sodium bicarbonate (500 mL) and the mixture was stirred at room temperature for 30 minutes. The organic layer was separated, dried over sodium sulfate, and the solvent was removed in vacuo to provide 25.8 g (99%) of 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide as a thick light yellow oil (crude).
  • Then to a solution of the crude 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide (as a free base) in methylene chloride (270 mL) at room temperature was added hydrogen chloride (65 mL, 1.0 M in ethyl ether) slowly. The resulting suspension was allowed to stir at room temperature for 1 hour. The solvent was removed in vacuo to afford 33 g as a yellow foam, mono hydrochloride salt. The foam was dissolved in solvents (330 mL, acetonitrile:methanol=33:1) at 60-70° C. and a crystal seed was added. The mixture was slowly cooled down to the room temperature and allowed to stir at room temperature for 15 hours. The resulting precipitate was filtered and dried to give 20.5 g (72%) of the title compound as a white crystal, mono hydrochloride salt. MS [M−H] m/z 412.3; mp. 132-133° C. 1H NMR (400 MHz, DMSO-d6) δ 10.6-10.8 (br, 1H), 10.45 (s, 1H), 7.64 (d, J=8 Hz, 2H), 7.00 (d, J=8 Hz, 2H), 6.74 (s, H), 4.00 (m, 1H), 3.77 (s, 3H), 3.4-3.6 (m, 6H), 2.9˜3.0 (m, 5H), 2.34 (m, 2H), 2.0 (s, 3H), 1.65-1.70 (m, 2H), 1.55-1.65 (m, 2H).
  • Example 3 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide (mono hydrochloride salt) i) 3-(4-methoxyphenyl)-3-oxopropanenitrile
  • A solution of methyl p-anisate in acetonitrile was cooled to −10° C. Lithium bis(trimethylsilyl)amide (1 M in THF) was added dropwise over a minimum of 3 hr. The mixture was held at −10 to 0° C. until reaction completion. The reaction mixture was quenched with water and the pH adjusted to 3-4 with conc HCl. The mixture was stirred for 1 hr. The product was isolated by filtration, washed with water and dried in a vacuum oven. The yield was 73%.
  • ii) 5-(4-methoxyphenyl)-1H-pyrazol-3-amine
  • A suspension of 3-(4-methoxyphenyl)-3-oxopropanenitrile in ethanol was heated to 60° C. Hydrazine hydrate was added dropwise over a minimum of 30 min at 60° C. The resulting solution was held at 60° C. until reaction completion, generally 15-18 hr. The reaction mixture was quenched with water. Ethanol was removed by distillation to about 5 volumes. The product was isolated by filtration, washed with water and dried in a vacuum oven. The yield was 88-95%.
  • iii) 5-bromo-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide
  • A solution of 5-(4-methoxyphenyl)-1H-pyrazol-3-amine and diisopropylethylamine in 10 volumes of a 9:1 mixture of acetonitrile:DMF was cooled to −10° C. 5-Bromovaleryl chloride was added dropwise over a minimum of 3 hr at −10° C. The resulting solution was held at −10° C. until reaction completion, generally 2 hr. The reaction mixture was quenched with water. The product was isolated by filtration, washed with water, TBME and suction dried. The product-wet cake was purified by re-slurrying in TBME at 35° C. for a minimum of 2 hr. The yield was 70-80%.
  • iv) 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide hydrochloride
  • Bromopyrazole is mixed with K2CO3 and KI in 10 volumes of acetone at room temperature and N-acetylhomopiperazine was added over 1 hr. The reaction mixture was stirred until the reaction was complete. The mixture was filtered, removing the inorganics, washed with acetone and distilled to 2 volumes. The freebase was extracted into methyl THF/EtOH and washed with NaCl and NaHCO3. The solvent was replaced with EtOH, a strength of the solution was determined, and 0.93 eq of HCl based on the available freebase was added to a mixture of acetone, ethanol and water. Careful monitoring of the pH yielded crystalline product in a 70% overall yield and the desired form 1.
  • Example 4 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide (mono hydrochloride salt) i) 5-(4-methoxy-phenyl)-1H-pyrazol-3-ylamine
  • The intermediate 5-(4-methoxy-phenyl)-1H-pyrazol-3-ylamine is commercially available from Sigma-Alrich (USA), but can be made using the following general procedure:
  • Aryl β-Ketonitrile Synthesis
  • To a solution of an aromatic ester (6.5 mmol) in dry toluene (6 mL), under N2, NaH (50-60% dispersion in mineral oil, 624 mg, 13 mmol) was carefully added. The mixture was heated at 80° C. and then dry CH3CN was added dropwise (1.6 mL, 30.8 mmol). The reaction was heated for 18 h and generally the product precipitated from the reaction mixture as a salt. The reaction was allowed to cool to room temperature and the solid formed was filtered and then dissolved in water. The solution was acidified with 2 N HCl solution, and upon reaching a pH between 2-4, the product precipitated and was filtered. If no precipitation occurred, the product was extracted with DCM. After aqueous workup, the products were generally pure enough to be used in the next step without further purification. The isolated yield was generally 40-80%.
  • Aryl Aminopyrazole Synthesis
  • To a solution of β-ketonitrile (7.5 mmol) in absolute EtOH (15 mL), hydrazine monohydrate (0.44 mL, 9.0 mmol) was added and the reaction was heated at reflux for 18 hrs. The reaction mixture was allowed to cool to room temperature and the solvent was evaporated under reduced pressure. The residue was dissolved in 20 mL of DCM and washed with water. The organic phase was concentrated to give a crude product that was purified by SiO2 column or by precipitation from Et2O. For example, the 2-methoxy derivative was purified by SiO2 chromatography, eluting with a DCM/MeOH gradient (from 100% DCM to 90/10 DCM/MeOH); the 3-methoxy derivative was triturated with Et2O. Yields were generally 65-90%.
  • ii) 5-bromo-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]amide
  • A solution of 5-bromovaleryl chloride (2.1 mL, 15.7 mmol) in dry dimethylacetamide (DMA) (35 mL) was cooled to −10° C. (ice water bath) under N2; a solution of 5-(4-methoxy-phenyl)-1H-pyrazol-3-ylamine (3.0 g, 15.7 mmol) and diisopropylethylamine (2.74 mL, 15.7 mmol) in dry DMA (15 mL) was added over 30 min. After two hours at −10° C., LCMS shows completion of the reaction (acylation on the pyrazole ring was also detected). The reaction was quenched by addition of H2O (ca. 50 mL), and the thick white precipitate formed upon addition of water was recovered by filtration. When the reaction was allowed to reach room temperature before quenching, a putative exchange of Br with Cl caused reactivity problems in subsequent steps. Washing with Et2O (3×10 mL) efficiently removed the byproduct (acylation on pyrazole ring). 4.68 g of the title compound was obtained as a white powder (13.3 mmol, 85% yield). Mp=149.5-151.5° C.
  • iii) 5-(4-acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide
  • 5-bromo-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]amide (1.5 g, 4.26 mmol) was dissolved in DMF (15 mL), and sodium iodide (0.64 g, 4.26 mmol) was added followed by N-acetylhomopiperazine (0.56 mL, 4.26 mmol) and diisopropylethylamine (0.74 mL, 4.26 mmol). The reaction was stirred under N2 at 50° C. for 18 hrs. Upon reaction completion (as monitored by LCMS), the solvent was removed at reduced pressure and the resulting oily residue was dissolved in DCM (20 mL), washed with sat. Na2CO3 (2×20 mL) and sat. NaCl (2×20 mL), and dried over Na2SO4. Upon solvent removal, 1.7 g of crude product as a thick oil were obtained. The product was purified by SiO2 chromatography (10 g cartridge-flash SI II from IST) employing DCM and DCM:MeOH 9:1 to yield 0.92 g of pure product and 0.52 g of less pure product. A second purification of the impure fractions using a 5 g SiO2 cartridge was performed using the same eluent. Overall, 1.09 g of 5-(4-acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide were obtained (2.64 mmol, 62% yield) as a thick light yellow oil. MS (ES+): 414.26 (M+H)+.
  • iv) 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide (mono hydrochloride salt)
  • 5-(4-acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide (1.05 g, 2.54 mmol) was dissolved in a minimum amount of DCM (5 mL) and cooled to 0° C. HCl (2.0 M in Et20, 1.4 mL, 2.89 mmol) was added and the mixture stirred at rt until precipitation of the salt was complete (about 10 min.). The solid was filtered, washed with Et2O several times, and dried in a dessicator to yield 1.09 g of the hydrochloride salt (2.42 mmol, 95% yield). Melting point was not determined due to the extreme hygroscopicity of the sample. MS (ES+): 414.26 (M+H)+.
  • Example 5 5-(4-acetyl-1,4-diazepan-1-yl)-N-(5-(4-methoxyphenyl)-1H-pyrazol-3-yl)pentanamide (mono hydrochloride salt) i) 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide
  • To a cylindrical, jacketed 3 L reactor equipped with nitrogen inerting, agitator, condenser/distillation head, and temperature control, 5-bromo-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]amide (0.15 kg, 0.426 mol), potassium carbonate (0.059 kg, 0.426 mol), potassium iodide (0.071 kg, 0.426 mol), and acetone (1.18 kg, 1.5 L) were added (at 20° C.) to form a white mixture. The mixture was stirred (235 rpm) at 25-30° C. for a minimum of 15 min. N-acetylhomopiperazine (0.062 kg, 0.057 L, 0.434 mol) was added via addition funnel to the reactor over a minimum of 45 min., maintaining the temperature in the range of 25-30° C. The addition funnel was rinsed with 0.05 L acetone. A white mixture persisted. The mixture was stirred (235 rpm) in the range of 25-30° C. for a minimum of 16 h, forming a white/yellow mixture. The reaction progress was monitored by HPLC and was considered complete when there was ≦2% of the starting material (bromopyrazole) and ≦2% of the iodopyrazole present.
  • The reactor contents were cooled to 5-15° C. over a minimum of 15 min with agitation (295 rpm) to form a white/yellow mixture that was then stirred for a minimum of 1 h. To remove inorganics, the mixture was then filtered on a Buchner funnel with filter paper using house vacuum for 1.5 min. The cake was washed twice with acetone (total of 0.24 kg, 0.30 L) at 5-15° C. The wash was combined with the mother liquor from the prior filtration and used to rinse the reactor. The filtrate was concentrated to a volume of approximately 0.45 L to form a clear solution.
  • ii) Aqueous Workup
  • To a reactor containing the material from step i, 1.5 L of a freshly made homogeneous solution of methyl THF (1.22 kg, 1.42 L) and ethanol (0.059 kg, 0.075 L; 99.5% ethanol, 0.5% toluene) was added at 25° C., forming a hazy solution. To this, 0.45 L of a 5% solution of sodium chloride (0.022 kg) in water (0.43 L) was added at 25° C. The resulting mixture was heated with stirring to 30-35° C. over a minimum of 15 min., forming a clear biphasic solution. The agitation was stopped to allow the layers to settle, the product being in the upper layer. The layers were separated, keeping any emulsion in the upper organic layer. The organic layer was retained. A homogeneous 5% solution of sodium bicarbonate (0.03 kg) in water (0.57 L) at 25° C. was used to wash organic layer, stirring for a minimum of 5 min. at 10-15° C. The agitation was stopped to allow the layers to settle, the product being in the upper layer. The layers were separated, keeping any emulsion in the upper organic layer. The organic layer was retained and concentrated to a volume of 0.35 L, forming a hazy solution. The mixture was chased with ethanol to remove residual water.
  • iii) 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide HCl
  • To a reactor containing the material from step ii, 0.47 kg (0.60 L) of acetone was added. The resulting mixture was heated with stirring to 25-30° C. over a minimum of 10 min., forming a hazy solution. The contents of the reactor were clarified through a polypropylene pad into a tared 2 L suction flask using vacuum, maintaining the contents of the reactor at 25-30° C. Suction was maintained until filtration stopped. The reactor and filter pad were rinsed with acetone (0.05 L) at 20-25° C. An accurate strength of the free base was determined. The filtrates from the suction flask were transferred to the reactor and rinsed using acetone (0.05 L). A solution of 5% HCl (0.042 kg, 0.036 L) in acetone (0.174 L) and alcohol solution (0.0174 L of ethanol:acetone (91:9) v/v) was prepared and stirred until homogeneous at 10° C. To the reactor, 0.05 L of water was added to form a clear solution. One third of the 5% HCl solution (0.076 L) was added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. A second third of the 5% HCl solution (0.076 L) was then added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. The contents of the reactor were seeded with 75 mg of 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide HCl (e.g., Form 1), followed by the addition of the last third of 5% HCl solution (0.076 L) over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. Another 0.08 equiv. of the 5% HCl solution (0.023 L) was then added to the reactor over a minimum of 30 min., maintaining the temperature in the range of 20-25° C. Judicious monitoring of pH was performed to attain the desired pH range of 5.2-5.8. Based on the strength calculation, 0.85 equiv. of acid was added over 1 hr and the remaining acid was added over a minimum of 30 min. with careful monitoring of pH.
  • The mixture was stirred at 20-25° C. for a minimum of 1 hr., forming a thin suspension. Acetone (0.6 L) was added over a minimum of 60 min., maintaining the temperature in the range of 20-25° C. The mixture was stirred at 20-25° C. for a minimum of 60 min. Acetone (1.5 L) was added to the reactor over a minimum of 3 hr., maintaining the temperature in the range of 20-25° C., forming a thick suspension. The mixture was then stirred at 20-25° C. for a minimum of 12 h. Crystallization was considered complete when there was ≦20% of the product present in the mother liquor. Longer stirring was employed if crystallization was not complete.
  • The mixture was then filtered on a Buchner funnel (polypropylene pad) using house vacuum. A solution of water (0.009 L), acetone (0.23 L) and 0.06 L alcohol (ethanol:acetone (91:9) v/v) was stirred until homogeneous (20% ethanol, 3% water, 77% acetone overall). This solution was used to wash the filter cake twice (0.15 L×2). A solution of water (0.009 L), acetone (0.171 L) and 0.12 L alcohol (ethanol:acetone (91:9) v/v) was stirred until homogeneous (40% ethanol, 3% water, 57% acetone overall). This solution was used to wash the filter cake (0.30 L). The wet cake was subjected to suction under nitrogen using house vacuum and held for 30 min. after dripping stopped. Product purity was checked by HPLC and additional washing was performed if total impurities were not ≦2%. Product was oven dried in a vacuum oven with nitrogen bleed at 38-45° C., maintaining vacuum at 20 torr for a minimum of 12 h until loss on drying of less than 1% was obtained. Following drying, 0.119 kg of the title compound was obtained in 62% yield (67% adjusted for aliquots removed during process; 60% when corrected for strength or purity). Melting point=185° C.; crystal form=form 1; particle size=D90<89.4 um, D50<19.2 um.
  • Example 6 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide (hydrochloride salt) crystal forms
  • The present Example describes the preparation of the hydrochloride salt form of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide. The hydrochloric acid salt form readily adopted a solid form. Indeed, at least four different crystalline forms (i.e., polymorphs) were observed for the hydrochloric acid salt form (see below).
  • Counter Ion Used Solid Obtained Melting Onset Hygroscopicity
    Hydrochloric acid Crystalline solid 185° C. No
    165° C. Somewhat
    125° C. Yes
    125° C. Not Measured
    three peaks: Yes
    about 100
    about 180; and
    about 200° C.
  • Differential scanning calorimetry data were collected for each solid form achieved using a DSC (TA instruments, model Q1000) under the following parameters: 50 mL/min purge gas (N2); scan range 40 to 200° C., scan rate 10° C./min. Thermogravimetric analysis data were collected using a TGA instruments (Mettler Toledo, model TGA/SDTA 851e) under the following parameters: 40 ml/min purge gas (N2); scan range 30 to 250° C., scan rate 10° C./min. X-ray data were acquired using an X-ray powder diffractometer (Bruker-axs, model D8 advance) having the following parameters: voltage 40 kV, current 40.0 mA, scan range 5 to 30°, scan step size 0.01°, total scan time 33 minutes, VANTEC detector, and antiscattering slit 1 mm. FIGS. 1-7 show characterization data for hydrochloride salt forms.
  • The hydrochloride salt was polymorphic, adopting crystalline forms exhibiting DSC endotherms at 119° C. (Form III), 127° C. (Form IV), 167° C. (Form II), and 186° C. (Form I). Another form, potentially an ethanol solvate/hydrate, exhibited multiple endotherms, corresponding to 1) desolvation at about 100° C., 2) Form I at about 183° C., and 3) possibly another polymorph at about 200° C. The Table below illustrates certain characteristics of observed hydrochloride salt crystal forms:
  • Crystal Form Table
    Crystal Form I
    Mono-
    hydrochloride Crystal Form Crystal Form
    (8% HCl) Crystal Form II III IV Crystal Form V
    Melting: 180-186° C. Melting: 165° C. Melting: 125° C. Melting: 125° C. Three peaks:
    About 100° C.
    About 180° C.
    About 200° C.
    Non-hygroscopic Somewhat Hygroscopic Not tested Hygroscopic (7%
    hygroscopic (5% (10% water at at RH 50%)
    water at RH RH 50%)
    50%)
  • Of the various observed hydrochloride forms, only Form I (186° C.) is relatively non-hygroscopic, gaining only about 0.5% moisture when equilibrated at RH less than or equal to 70%. At 70-100% RH, Form I gains at least about 2% moisture, but loses it without significant hysteresis on decreasing RH. Evidence of a hydrochloride hydrate was not observed after the hygroscopicity test.
  • Higher degrees of hydrochloride salt were formed, depending on the amount of hydrochloric acid present in the solution during reactive crystallization. The conversion of higher degrees of hydrochloride salt to mono-hydrochloride salt can be achieved by adjusting the pH of the solution to more than pH 5. Further adjustment, however, can result in formation of inorganic salts. In some embodiments, pure mono-hydrochloride salt forms are produced with hydrochloride equivalence and slurry pH of <0.95 eq. (e.g., 0.93) and pH 5, respectively (see, for example, FIGS. 8-11).
  • Example 7 Characterization of Certain Crystal Forms of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-amide hydrochloride Salt
  • The present Example describes characterization of two surprisingly non-hygroscopic crystal forms (Forms I and II, as described above) of a hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide:
  • Figure US20090264648A1-20091022-C00044
  • Both forms are considerably soluble in water. The melting point of Form I is 185° C. (plus or minus 2 degrees); the melting point of Form II is 166° C. (plus or minus 5 degrees).
  • Form I picks up moisture at relative humidity (RH) of about 50% and absorbs up to about 2% water eventually (90% RH) and loses the water as RH decreases (<50%). Form I also exhibits characteristic X-ray peaks at 20 of 15.3° and 21.9°, plus or minus about 0.3°, depending upon the machine and measurement method utilized.
  • Form II picks up moisture at RH of about 20% and absorbs up to 7% water eventually (RH of 90%) and holds 2% at low RH (0%). Form II also exhibits characteristic X-ray peaks at 2θ of 20.2° and 24.9°, plus or minus about 0.3°, depending upon the machine and measurement method utilized. Differential scanning calorimetry data were collected for each solid form achieved using a DSC (TA instruments, model Q1000) under the following parameters: 50 mL/min purge gas (N2); scan range 40 to 200° C., scan rate 10° C./min.
  • Thermogravimetric analysis data were collected using a TGA instruments (Mettler Toledo, model TGA/SDTA 851e) under the following parameters: 40 ml/min purge gas (N2); scan range 30 to 250° C., scan rate 10° C./min.
  • X-ray data were acquired using an X-ray powder diffractometer (Bruker-axs, model D8 advance) having the following parameters: voltage 40 kV, current 40.0 mA, scan range (2θ) 3.7 to 30°, scan step size 0.0°, total scan time 33 minutes, VANTEC detector, and antiscattering slit 1 mm.
  • Dynamic Vapor Sorption (DVS) was done at 25-26° C.
  • Results of thermal studies on Crystal Forms I and II are shown in the ensuing Figures.
  • Example 8 Preparation of Crystal Form I of the Hydrochloride Salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide
  • The present Example describes the preparation of crystal form I of the hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide.
  • First procedure: 611.7 mg of the free base form of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide was dissolved in 1.97 mL acetone at 35° C. A solution of 5% HCl in acetone-water was prepared by diluting 37.5% aq. HCl using acetone. 0.6 ml of 5% HCl was added slowly. 1.2 ml EtOH ASDQ (100:10 ethanol:methanol) was added slowly. The solution became milky in a few minutes; stirring was performed for around 5 minutes. 0.25 ml of 5% HCl was added slowly. After 5 minutes, 0.25 ml of 5% HCl was added slowly. After 5 minutes, 0.087 ml of 5% HCl was added slowly. The mixture was heated to about 40-50° C. The mixture was left at room temperature while stirring overnight. Crystals were filtered and washed with 2 ml acetone, and were dried at 45° C. for about 7 hours. 505 mg of solid were recovered.
  • Second procedure: 377 mg of the free base form of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide was dissolved in 1.2 ml acetone at 35° C. 0.754 ml ethanol ASDQ (100:10 ethanol:methanol) was added. A solution of 5% HCl in acetone-water was prepared by diluting 37.5% aq HCl using acetone. 0.18 ml diluted HCl solution was added slowly. A seed of crystal form I of the hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide was added. 0.18 ml diluted HCl solution was added slowly. Around two minutes later, 0.18 ml diluted HCl solution was added slowly. Around two minutes later, another 0.18 ml diluted HCl solution was added slowly. The mixture was heated to about 40-50° C., and then was left at room temperature while stirring overnight. The crystals were filtered and washed with 1.5 ml acetone, and were dried at 45° C. for about 6 hours.
  • Example 9 Alternative purification of the hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide
  • The hydrochloride salt of 5-(4-Acetyl-[1,4]diazepan-1-yl)-pentanoic acid [5-(4-methoxy-phenyl)-2H-pyrazol-3-yl]-amide (compound I) may be re-purified by basifying the hydrochloride salt and extracting the free base into a suitable solvent (e.g. methylene chloride). The organic extracts may be washed with water. The organic phase is concentrated and the solvent switched to ethanol. Acetone is added to give a solution of compound A which was clarified and mixed with ethanol, acetone, hydrochloric acid, and water. Acetone is added, the solids filtered, washed with mixture of acetone, water and dried to give compound I. A representative procedure is described below.
  • To a suitable reactor, compound I (0.2 kg) was dissolved in water (0.80 L) and clarified through a filter pad. To the filtrates was added methylene chloride (2.65 kg) and cooled to 15° C. A 30% aqueous solution of sodium hydroxide (0.062 kg) was added over 30 mins and mixed for 20 mins. The pH was >8. The layers were separated; the organic layer was washed with water (2×0.40 kg) and distilled down to 0.46 L forming a hazy mixture. The methylene chloride solvent was exchanged with ethanol by vacuum distillation chases (2×0.79 kg).
  • Acetone (0.63 kg) was added to the concentrate and the solution clarified. An accurate strength of the free base was determined of the concentrate. Water (0.065 kg) was added to form a clear solution. A solution of 5% HCl (0.043 kg) in acetone (0.14 kg) and alcohol (0.14 kg of ethanol:acetone (91:9) v/v) was prepared and stirred until homogeneous at 10° C. About one third of the 5% HCl solution (0.098 kg) was added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. A second third of the 5% HCl solution (0.098 kg) was then added to the reactor over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. The contents of the reactor were seeded with 75 mg of 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide HCl (e.g., Form 1), followed by the addition of the last third of 5% HCl solution (0.098 kg) over a minimum of 20 min., maintaining the temperature in the range of 20-25° C. The contents of the reactor were seeded with another 75 mg of 5-(4-acetyl-[1,4]diazepan-1-yl)-N-[5-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-pentanamide HCl (e.g., Form 1). Another 0.08 equiv. of the 5% HCl solution (0.029 kg) was then added to the reactor over a minimum of 30 min., maintaining the temperature in the range of 20-25° C. Judicious monitoring of pH was performed to attain the desired pH range of 5.2-5.8. The mixture was stirred at 20-25° C. for a minimum of 1 hr., forming a thin suspension. Acetone (0.63 kg) was added over a minimum of 60 min., maintaining the temperature in the range of 20-25° C. The mixture was stirred at 20-25° C. for a minimum of 60 min. Acetone (1.58 kg) was added to the reactor over a minimum of 3 hr., maintaining the temperature in the range of 20-25° C., forming a thick suspension. The mixture was then stirred at 20-25° C. for a minimum of 12 h. Crystallization was considered complete when there was <15% of the product present in the mother liquor. Longer stirring was employed if crystallization was not complete. The mixture was then filtered on a Buchner funnel (polypropylene pad) using house vacuum. A solution of water (0.012 kg), acetone (0.24 kg) and 0.063 kg alcohol (ethanol:acetone (91:9) v/v) was stirred until homogeneous (20% ethanol, 3% water, 77% acetone overall). This solution was used to wash the filter cake. A solution of water (0.012 kg), acetone (0.18 kg) and 0.13 kg alcohol (ethanol:acetone (91:9) v/v) was stirred until homogeneous (40% ethanol, 3% water, 57% acetone overall). This solution was used to wash the filter cake. The wet cake was subjected to suction under nitrogen using house vacuum and held for 30 min. after dripping stopped. Product purity was checked by HPLC and additional washing was performed if total impurities were not ≦2%. Product was oven dried in a vacuum oven with nitrogen bleed at 38-45° C., maintaining vacuum at 20 torr for a minimum of 12 h until loss on drying of less than 1% was obtained. Following drying, 0.17 kg of the title compound was obtained in 85% yield.

Claims (34)

1. A method comprising the steps of:
providing a compound of formula I-1:
Figure US20090264648A1-20091022-C00045
wherein:
Ring A is a 4-8 membered saturated ring, having 0-2 heteroatoms independently selected from O, N, or S in addition to the nitrogen depicted in Ring A, wherein Ring A is independently substituted with 0-4 R′ groups;
R′ is selected from the group consisting of mono- or di-[linear, branched or cyclic C1-6 alkyl]aminocarbonyl; linear, branched or cyclic C1-6 alkyl, alkoxy, or acyl;
Y and Y′ are each independently N or C, with the proviso that at least one of Y or Y′ is N;
T is a C3-5 bivalent hydrocarbon chain, optionally carrying an oxo group and optionally substituted with one or more halogen, hydroxy, C1-5 alkyl, alkoxy, fluoroalkyl, hydroxyalkyl, alkylidene, or fluoroalkylidene groups; C3-6 cycloalkane-1,1-diyl, oxacycloalkane-1,1-diyl, C3-6 cycloalkane-1,2-diyl, or oxacycloalkane-1,2-diyl groups, wherein the bonds of the 1,2-diyl radical form a fused ring with the T chain; and with the proviso that when T carries an oxo group, said oxo group is not part of an amide bond; and
Ar is a group selected from 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur; wherein Ar is optionally substituted with one or more substituents independently selected from halogen; hydroxy; mercapto; cyano; nitro; amino; sulfonyl; linear, branched or cyclic (C1-C6) alkyl, trihaloalkyl, di- or trihaloalkoxy, alkoxy, or alkylcarbonyl; (C3-C6) cycloalkyl-(C1-C6) alkoxy; (C3-C6) cycloalkyl-(C1-C6) alkyl; linear, branched, or cyclic (C1-C6) alkylcarbonylamino; mono- or di-, linear, branched, or cyclic (C1-C6) alkylaminocarbonyl; carbamoyl; linear, branched, or cyclic (C1-C6) alkylsulphonylamino; linear, branched, or cyclic (C1-C6) alkylsulphonyl; mono- or di-, linear, branched, or cyclic (C1-C6) alkylsulphamoyl; linear, branched or cyclic (C1-C6) alkoxy-(C1-C6) alkyl; wherein, two substituents may be taken together with their intervening atoms to form a ring; and
(b) treating said compound of formula I-1 with hydrochloric acid in a ternary solvent solvent system to form a compound of formula I-1′:
Figure US20090264648A1-20091022-C00046
2. The method according to claim 1, wherein the ternary solvent system comprises acetone, water, and ethanol.
3. The method according to claim 2, wherein the hydrochloric acid is added as about a 5% solution in acetone and ethanol to a compound of formula I-1 in a mixture of acetone and water.
4. The method according to claim 3, wherein about 0.93 equivalents of hydrochloric acid is added relative to a compound of formula I-1.
5. The method according to claim 1, further comprising the steps of:
(c) providing compound of formula B′:
Figure US20090264648A1-20091022-C00047
wherein, LG2 is a suitable leaving group, and
(d) treating said compound of formula B′ with a compound of formula G′:
Figure US20090264648A1-20091022-C00048
optionally in the presence of a suitable base and/or additive, to form compound I-1.
6. The method according to claim 5, wherein LG2 is selected from Br, I, OMs, OTs, or OTf.
7. The method according to claim 6, wherein LG2 is Br.
8. The method according to claim 5, further comprising the steps of:
(e) providing a compound of formula C′:
Figure US20090264648A1-20091022-C00049
and
(f) treating said compound of formula C′ in the presence of a suitable base with a compound of formula F′:
Figure US20090264648A1-20091022-C00050
wherein LG is a suitable leaving group, to form a compound of formula B′.
9. The method of claim 1, wherein the compound of formula I-1 is compound A:
Figure US20090264648A1-20091022-C00051
10. The method of claim 1, wherein the compound of formula I-1′ is compound I:
Figure US20090264648A1-20091022-C00052
11. The method of claim 9, wherein the hydrochloric acid is added as about a 5% solution in acetone and ethanol to a compound of formula A in a mixture of acetone and water.
12. The method according to claim 11, wherein about 0.93 equivalents of hydrochloric acid is added relative to a compound A.
13. The method of claim 5, wherein the compound of formula B′ is:
Figure US20090264648A1-20091022-C00053
14. The method according to claim 13, wherein LG2 is selected from Br, I, OMs, OTs, or OTf.
15. The method according to claim 14, wherein LG2 is Br.
16. The method according to claim 13, wherein the base is pyridine, diisopropylethylamine, triethylamine, sodium bicarbonate, sodium carbonate, potassium carbonate, or combinations thereof.
17. The method according to claim 16, wherein the base is diisopropylethylamine.
18. The method according to claim 16, wherein the base is potassium carbonate.
19. The method according to claim 13, wherein the additive is an iodide source selected from sodium iodide, potassium iodide, hydrogen iodide, tetralkylammonium iodide, or a mixture thereof.
20. The method according to claim 19, wherein the iodide source is potassium iodide.
21. The method according to claim 19, wherein the iodide source is sodium iodide.
22. The method of claim 8, wherein the compound of formula C′ is:
Figure US20090264648A1-20091022-C00054
and the compound of formula F′ is:
Figure US20090264648A1-20091022-C00055
23. The method according to claim 22, wherein LG3 is selected from halogen, OR,
Figure US20090264648A1-20091022-C00056
wherein each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
24. The method according to claim 23, wherein LG3 is Cl.
25. The method according to claim 22, wherein LG2 is halogen.
26. The method according to claim 22, wherein a compound of formula F is selected from 5-bromovaleryl chloride or 5-iodovaleryl chloride.
27. The method according to claim 26, wherein a compound of formula F is 5-bromovaleryl chloride.
28. The method of claim 22, further comprising the steps of:
(g) providing compound D:
Figure US20090264648A1-20091022-C00057
and
(h) treating said compound D with hydrazine, or an equivalent thereof, to form compound
C:
Figure US20090264648A1-20091022-C00058
29. The method of claim 28, further comprising the steps of:
(a) providing a compound of formula E:
Figure US20090264648A1-20091022-C00059
wherein, LG1 is a leaving group, and
(b) treating said compound of formula with acetonitrile to form a mixture thereof, and
(c) treating said mixture with a suitable base to give compound D:
Figure US20090264648A1-20091022-C00060
30. The method according to claim 29, wherein LG1 is a halogen, alkoxy, sulphonyloxy, optionally substituted alkylsulphonyl, optionally substituted alkenylsulfonyl, optionally substituted arylsulfonyl, or diazonium moiety.
31. The method according to claim 30, wherein LG1 is methoxy.
32. A method comprising the steps of:
(a) providing a compound of formula C′:
Figure US20090264648A1-20091022-C00061
and
(b) treating said compound of formula C′ in the presence of a suitable base with a compound of formula F′:
Figure US20090264648A1-20091022-C00062
wherein LG2 and LG3 are suitable leaving groups, to form a compound of formula B′:
Figure US20090264648A1-20091022-C00063
and
(c) treating said compound of formula B′ with a compound of formula G′:
Figure US20090264648A1-20091022-C00064
optionally in the presence of a suitable base and/or additive, to form compound I-1:
Figure US20090264648A1-20091022-C00065
and
(d) treating said compound of formula I-1 with hydrochloric acid in a ternary solvent solvent system to form a compound of formula I-1′:
Figure US20090264648A1-20091022-C00066
wherein Ring A is a 4-8 membered saturated ring, having 0-2 heteroatoms independently selected from O, N, or S in addition to the nitrogen depicted in Ring A, wherein Ring A is independently substituted with 0-4 R′ groups;
R′ is selected from the group consisting of mono- or di-[linear, branched or cyclic C1-6 alkyl]aminocarbonyl; linear, branched or cyclic C1-6 alkyl, alkoxy, or acyl;
Y and Y′ are each independently N or C, with the proviso that at least one of Y or Y′ is N;
T is a C3-5 bivalent hydrocarbon chain, optionally carrying an oxo group and optionally substituted with one or more halogen, hydroxy, C1-5 alkyl, alkoxy, fluoroalkyl, hydroxyalkyl, alkylidene, or fluoroalkylidene groups; C3-6 cycloalkane-1,1-diyl, oxacycloalkane-1,1-diyl, C3-6 cycloalkane-1,2-diyl, or oxacycloalkane-1,2-diyl groups, wherein the bonds of the 1,2-diyl radical form a fused ring with the T chain; and with the proviso that when T carries an oxo group, said oxo group is not part of an amide bond; and
Ar is a group selected from 6-10 membered aryl, or 5-10 membered heteroaryl having 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur; wherein Ar is optionally substituted with one or more substituents independently selected from halogen; hydroxy; mercapto; cyano; nitro; amino; sulfonyl; linear, branched or cyclic (C1-C6) alkyl, trihaloalkyl, di- or trihaloalkoxy, alkoxy, or alkylcarbonyl; (C3-C6) cycloalkyl-(C1-C6) alkoxy; (C3-C6) cycloalkyl-(C1-C6) alkyl; linear, branched, or cyclic (C1-C6) alkylcarbonylamino; mono- or di-, linear, branched, or cyclic (C1-C6) alkylaminocarbonyl; carbamoyl; linear, branched, or cyclic (C1-C6) alkylsulphonylamino; linear, branched, or cyclic (C1-C6) alkylsulphonyl; mono- or di-, linear, branched, or cyclic (C1-C6) alkylsulphamoyl; linear, branched or cyclic (C1-C6) alkoxy-(C1-C6) alkyl; wherein, two substituents may be taken together with their intervening atoms to form a ring;
wherein each step is performed sequentially without isolation of intermediates B′ or I-1.
33. The method according to claim 32, wherein the compound of formula F′ is slowly added to the compound of formula C′ in step (b).
34. The method according to claim 33, wherein about 0.5-0.95 equivalents of hydrochloric acid is added relative a compound of formula I-1.
US12/353,896 2008-01-14 2009-01-14 Synthesis of pyrazoles Abandoned US20090264648A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/353,896 US20090264648A1 (en) 2008-01-14 2009-01-14 Synthesis of pyrazoles

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US2101508P 2008-01-14 2008-01-14
US12/353,896 US20090264648A1 (en) 2008-01-14 2009-01-14 Synthesis of pyrazoles

Publications (1)

Publication Number Publication Date
US20090264648A1 true US20090264648A1 (en) 2009-10-22

Family

ID=40613075

Family Applications (1)

Application Number Title Priority Date Filing Date
US12/353,896 Abandoned US20090264648A1 (en) 2008-01-14 2009-01-14 Synthesis of pyrazoles

Country Status (2)

Country Link
US (1) US20090264648A1 (en)
WO (1) WO2009091832A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100029606A1 (en) * 2007-01-16 2010-02-04 Wyeth Modulators of alpha7 nicotinic acetylcholine receptors and therapeutic uses thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105646358B (en) * 2016-03-01 2018-03-09 苏州艾缇克药物化学有限公司 A kind of synthetic method of 3 amino 5 (4 anisyl) pyrazoles
IT201600092469A1 (en) * 2016-09-14 2018-03-14 Lundbeck Pharmaceuticals Italy S P A Blonanserine production process

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2637530A1 (en) * 2006-01-18 2007-09-07 Siena Biotech S.P.A. Modulators of alpha7 nicotinic acetylcholine receptors and therapeutic uses thereof
TW200901974A (en) * 2007-01-16 2009-01-16 Wyeth Corp Compounds, compositions, and methods of making and using them

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100029606A1 (en) * 2007-01-16 2010-02-04 Wyeth Modulators of alpha7 nicotinic acetylcholine receptors and therapeutic uses thereof
US8163729B2 (en) 2007-01-16 2012-04-24 Wyeth Modulators of α7 nicotinic acetylcholine receptors and therapeutic uses thereof

Also Published As

Publication number Publication date
WO2009091832A1 (en) 2009-07-23

Similar Documents

Publication Publication Date Title
US8884016B2 (en) Apixaban preparation process
US7863460B2 (en) Process for producing 1-substituted-3-fluoroalkylpyrazole-4-carboxylate
US9643927B1 (en) Process for the preparation of kinase inhibitors and intermediates thereof
US10550087B2 (en) Process for the preparation of kinase inhibitors and intermediates thereof
JP7446316B2 (en) Substituted pyrrolidine amide III
JP6268093B2 (en) Process for producing fused heterocyclic derivative and production intermediate thereof
JP4330836B2 (en) Process for producing piperazine derivatives
US20090264648A1 (en) Synthesis of pyrazoles
EP3413891A1 (en) Processes for the preparation of highly pure prucalopride succinate and its intermediates
EP2556064B1 (en) Process for the preparation of 2-(cyclohexylmethyl)-N-{2-[(2S)-1-methylpyrrolidin-2-yl]ethyl}-1,2,3,4-tetrahydroisoquinoline-7-sulfonamide
US20060069270A1 (en) Process for the preparation of 1,3,5-trisubstituted pyrazoles via [3+2] cycloaddition
US6800758B1 (en) Process for the preparation of a 3(2H)-pyridazinone-4- substituted amino 5-chloro derivative
US20120041211A1 (en) Novel process for preparing carboxy-containing pyrazoleamido compounds 597
US8815870B2 (en) 4-(2-(6-substituted-hexylidene) hydrazinyl)benzonitrile and preparation thereof
JP4238978B2 (en) Benzazepine compounds and process for producing the same
US20210147401A1 (en) Process for the production of 5-(4-((2s,5s)-5-(4-chlorobenzyl)-2-methyl-morpholino)piperidin-1-yl)-1h-1,2,4-triazol-3-amine
CA3214107A1 (en) New process for the synthesis of 5-{5-chloro-2-[(3s)-3- [(morpholin-4-yl)methyl]-3,4-dihydroisoquinoline-2(1h)- carbonyl]phenyl}-1,2-dimethyl-1h-pyrrole-3-carboxylic acid derivatives and its application for the production of pharmaceutical compounds
KR100377578B1 (en) Process for the preparation of ondansetron and pharmaceutically acceptable salts thereof
EP1829870A1 (en) 2-(pyrazol-1-yl)pyridine derivative
JP2020529412A (en) Intermediate compounds and methods
JP2000186081A (en) Production of methylamine derivative
JPH07247275A (en) Production of single diastereomer of oxazolidinone derivative and fumarate of single diastereomer of urethane compound
JP2009046400A (en) Production intermediate for insecticidal compound and method for producing the same

Legal Events

Date Code Title Description
AS Assignment

Owner name: SIENA BIOTECH S.P.A., ITALY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NENCINI, ARIANNA;ZANALETTI, RICCARDO;REEL/FRAME:022226/0954;SIGNING DATES FROM 20090130 TO 20090202

AS Assignment

Owner name: WYETH, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEW, WARREN;WANG, ZHENG;CHEAL, GLORIA;AND OTHERS;REEL/FRAME:022920/0507;SIGNING DATES FROM 20090318 TO 20090528

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION